WO2018160849A1 - Thérapie génique pour troubles oculaires - Google Patents

Thérapie génique pour troubles oculaires Download PDF

Info

Publication number
WO2018160849A1
WO2018160849A1 PCT/US2018/020470 US2018020470W WO2018160849A1 WO 2018160849 A1 WO2018160849 A1 WO 2018160849A1 US 2018020470 W US2018020470 W US 2018020470W WO 2018160849 A1 WO2018160849 A1 WO 2018160849A1
Authority
WO
WIPO (PCT)
Prior art keywords
lca5
raav
aav
capsid
promoter
Prior art date
Application number
PCT/US2018/020470
Other languages
English (en)
Other versions
WO2018160849A8 (fr
Inventor
Jean Bennett
Jeannette BENNICELLI
Junwei Sun
Ji-yun SONG
Sergei NIKONOV
Original Assignee
The Trustees Of The University Of Pennsylvania
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP18760861.7A priority Critical patent/EP3589738A4/fr
Priority to BR112019017327A priority patent/BR112019017327A2/pt
Application filed by The Trustees Of The University Of Pennsylvania filed Critical The Trustees Of The University Of Pennsylvania
Priority to AU2018228881A priority patent/AU2018228881B2/en
Priority to KR1020197027823A priority patent/KR102545070B1/ko
Priority to JP2019547457A priority patent/JP7211960B2/ja
Priority to CN201880029279.2A priority patent/CN110582572A/zh
Priority to KR1020237020037A priority patent/KR20230093072A/ko
Priority to US16/489,770 priority patent/US11564996B2/en
Priority to RU2019130004A priority patent/RU2019130004A/ru
Priority to CA3054136A priority patent/CA3054136A1/fr
Publication of WO2018160849A1 publication Critical patent/WO2018160849A1/fr
Priority to IL26894619A priority patent/IL268946A/en
Publication of WO2018160849A8 publication Critical patent/WO2018160849A8/fr
Priority to US18/061,633 priority patent/US20230233709A1/en
Priority to JP2023002799A priority patent/JP2023040219A/ja
Priority to AU2024202693A priority patent/AU2024202693A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14145Special targeting system for viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/22Vectors comprising a coding region that has been codon optimised for expression in a respective host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/50Vector systems having a special element relevant for transcription regulating RNA stability, not being an intron, e.g. poly A signal

Definitions

  • LCA Leber congenital amaurosis
  • OMIM 204000 One of the most severe groups of inherited blinding diseases is Leber congenital amaurosis (LCA; OMIM 204000).
  • LCA is rare, occurring in 1 :50,000 individuals, is usually inherited in an autosomal recessive fashion, and can be caused by mutations in any of at least 22 different genes (sph.uth.edu/RetNet/sum-dis.htm).
  • Clinical features include severely abnormal vision (visual acuity, reduced visual fields) in infancy or early childhood, nystagmus, and progressive loss of the poor vision that exists early in life.
  • Clinical testing reveals extinguished scotopic and photopic electroretinogram (ERG) responses, amaurotic pupils, reduced light sensitivity, and pigmentary changes in the retina. There is currently no approved treatment for LCA.
  • ERP photopic electroretinogram
  • RPE65 Redmond TM, Yu S, Lee E, Bok D, Hamasaki D, Chen N, et al. Rpe65 is necessary for production of 1 1-cis-vitamin A in the retinal visual cycle. Nat Genet (1998) 20(4):344-51 ; Redmond T, Hamel C. Genetic analysis of RPE65 : from human disease to mouse model. Methods in Enzymol (2000) 317:705-24, both of which are incorporated herein by reference), has received a great deal of attention in recent years as it has been the target of multiple gene augmentation therapy clinical trials.
  • AAV adeno-associated virus serotype 2
  • RPE retinal pigment epithelium
  • LCA5 One of the most severe forms of this already severe condition (LCA) is caused by mutations in the photoreceptor-specific gene encoding Lebercilin, LCA5 (12-20). LCA5 mutation is estimated to account for ⁇ 2% of cases of LCA although it may be more prevalent in populations that are genetically isolated. (16) LCA5 mutations have also been identified as the cause of other early onset forms of retinal degeneration, including cone dystrophy and autosomal recessive retinitis pigmentosa (ARRP).(15, 16, 21)
  • ARRP autosomal recessive retinitis pigmentosa
  • compositions useful for expressing Lebercilin in subjects in need are needed.
  • LCA Leber congenital amaurosis
  • a codon optimized, engineered nucleic acid sequence encoding human Lebercilin is provided.
  • the codon optimized nucleic acid sequence is a variant of SEQ ID NO: 3 or SEQ ID No: 2.
  • the codon optimized nucleic acid sequence is SEQ ID NO: 3.
  • the nucleic acid sequence is codon optimized for expression in humans.
  • an expression cassette comprising a codon optimized nucleic acid sequence that encodes Lebercilin.
  • the expression cassette includes the nucleic acid sequence of SEQ ID NO: 3 encoding human Lebercilin.
  • the Lebercilin encoding sequence is positioned between 5' and 3' AAV ITR sequences.
  • a recombinant adeno-associated virus (rAAV) vector is provided. The rAAV compromises an AAV capsid, and a vector genome packaged therein.
  • the vector genome comprises: (a) an AAV 5' inverted terminal repeat (ITR) sequence; (b) a promoter; (c) a coding sequence encoding a human Lebercilin; and (d) an AAV 3' ITR.
  • the rAAV vector further comprises expression control sequences that direct expression of the Lebercilin in a host cell.
  • the Lebercilin sequence is the protein sequence of SEQ ID NO: 1.
  • the vector genome is the sequence of nt 1-4379 of SEQ ID NO: 8.
  • the vector genome is the sequence of nt 1-4368 of SEQ ID NO: 9.
  • the LCA5 coding sequence in either of the identified vector genomes is swapped with another LCA5 coding sequence as described herein.
  • an aqueous suspension suitable for administration to an LCA patient comprises an aqueous suspending liquid and about 1 xlO 10 GC or viral particles to about 1 xlO 13 GC or viral particles per eye of a recombinant adeno-associated virus (rAAV) described herein useful as a therapeutic for LCA.
  • rAAV recombinant adeno-associated virus
  • a pharmaceutical composition comprises a pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant and the nucleic acid sequence, a plasmid, a vector, or a viral vector, such as the rAAV, described specifically herein.
  • a method for treating Leber Congenital Amaurosis caused by a defect in the lebercilin gene (LCA5) and/or restoring visual function in a mammalian subject having LCA comprises delivering via intravitreal, subretinal or intravascular injection to a subject in need thereof a recombinant AAV vector which encodes Lebercilin, as described herein.
  • an AAV vector as described herein in treating Leber Congenital Amaurosis caused by a defect in the lebercilin gene (LCA5) and/or restoring visual function in a mammalian subject having LCA.
  • the use includes delivering via intravitreal, subretinal or intravascular injection to a subject in need thereof a recombinant AAV vector which encodes Lebercilin, as described herein.
  • FIG 1A shows the transgene cassettes used to generate AAV7m8.CBA.hopt.LCA5 and AAV7m8.CBA.EGFP as described herein.
  • FIGs IB-ID provide an alignment of human nucleic acid sequence of LCA5
  • FIGs 1E- 1F provide a plasmid map and a feature list of the pAAV.CMV.CBA.human codon-optimized Lebercilin vector.
  • the nucleic acid sequence is reproduced in SEQ ID NO: 8.
  • FIGs 1G-1H provide immunofluorescence analysis of eyes injected with AAV7m8-hopt- LCA5 at P5 and analyzed at P 15 shows Lebercilin co-localizing with the base of tubulin-positive outer segments as described in Examples 1, 2 and 4.
  • IVT intravitreal injection
  • SR subretinal injection
  • Lebercilin was distributed throughout the retina, which was nearly devoid of photoreceptors at P95.
  • lebercilin is absent in untreated P 15 and P95 Lca5-/- retinas.
  • SR subretinal
  • IVT intravitreal
  • Cartoons areprovided showing the intravitreal injection scheme (FIG 1G) and subretinal injection scheme (FIG 1H).
  • FIG II shows expression of Lebercilin in both wild type mouse and Lca5-/- mice at P20 injected with the AAV.LCA5 vector subretinally.
  • FIGs 2A-2I show normalized pupillary reflex amplitudes measured at 3 months of age of
  • FIGs 2F and 2G are schematic representations of the relative pupillary reflex amplitudes (% of baseline pupil diameter) of the right eyes of animals in each of the sub-groups shown in (A-C) plus in wildtype (C57B16) positive (+) control mice.
  • FIGs 2H and 21 show a comparison of normalized pupillary reflex amplitudes in experimental and control mice, shown in FIGs 2A-2D, treated at PN5 vs PN15 via subretinal (SR, I) or intravitreal (IV, H) injection. * . p ⁇ 0.1 ; ** p ⁇ 0.05; * **p ⁇ 0.01.
  • FIGs 3A-3F show results of water maze test AAV7m8.hopt-LCA5-injected vs. control
  • FIG. 3A is a table showing the statistical analysis result.
  • FIG. 3B is an illustration of representative results of water maze test.
  • FIG. 3D is a bar graph showing days until first success in training of Lca5-/- mice at PN5 or PN15 treated at birth with the AAV7m8.LCA5 vector intravitreally or subretinally at birth. Wild type mice were provided as controls.
  • FIG. 3D is a line graph of success rates under various light intensities (x- axis) of Lca5-/- mice at PN5 treated at birth with the AAV7m8.LCA5 vector intravitreally.
  • FIG. 3E is a line graph of success rates under various light intensities (x- axis) of Lca5-/- mice at PN5 treated at birth with the AAV7m8.LCA5 vector subretinally.
  • FIG. 3F is a line graph of success rates under various light intensities (x- axis) of Lca5-/- mice at PN15 treated at birth with the AAV7m8.LCA5 vector intravitreally.
  • FIG. 4 is a graph providing results of representative histology after AAV7m8.hopt-LCA5 delivery to the Lca5gt/gt retina at postnatal day (PN)5.
  • the graph shows the number of rows in the outer nuclear layer (ONL) after IVT or SR treatment with AAV.LCA5 vs. sham-injection.
  • FIG 5 A provides result of immunofluorescence shows that rhodopsin (red) persists in the treated (but not control untreated) photoreceptors through the 3 month timepoint.
  • Occasional photoreceptor cells are eGFP-positive (region of injection identified through co-injection with AAV7m8.eGFP).
  • FIGs. 5B-5D provide Multi-electrode array (MEA) responses from Lca5gt/gt
  • AAV7m8.hopt-LCA5 -treated rods and cones are similar to those of wild type (WT) retinas.
  • B Response amplitude (difference in firing rates before and after flash onset) vs. flash intensity data measured from per flash averaged traces (not shown). Responses from treated retinas are as high as 70% of the response from WT retina; there is minimal to abolished response from untreated retina.
  • C Response amplitudes from retinas of panel A for the 1st and 2nd intensity series runs (before/during and after brightest exposures at the end of the 1st run, during each intensity series run stimulation series intensity was increased from scotopic to the brightest photopic values in -0.5 log increments)
  • D Amplitudes of transient ON- (difference in firing rates before and after flash onset), sustained ON- (difference before flash onset and offset) and OFF-responses
  • FIGs 6A-6E show that there is massive cell death during photoreceptor degeneration early in life in the untreated Lca5-/- retina (and delay of this degeneration after treatment with
  • FIGs 7A-7D show that outer segments were present in AAV7m8.hopt.LCA5-treated Lca5-/- retinas but not in control Lca5-/- retinas. Transmission electron microscopic evaluation of the retinas injected with AAV7m8.hopt.LCA5 reveals both rod and cone photoreceptor outer segments with stacked discs and connecting cilia in 3 month old Lca5-/-. No such structures were present in untreated Lca5-/- retinas. (A, B) PN80 after intravitreal injection at PN5 with
  • AAV7m8p643 codon optimized Lebercilin, representative pictures; (A 1-A3): 12K resolution, stitched pictures showing rows of phororeceptor cells, Rod cells (arrow heads), cone cells (arrows), many mitochondria of photoreceptor cells (asterisks); (B), 20K resolution; (B l) cross section of cilia showing 9+0 structure of microtubule, membraneous disc of OS from Rod cell (arrows); (C) 30K resolution, basal body (arrow head); (C I) sagital section of connecting cilium; (D) 12K resolution, no photoreceptor cells remained in ONL; (Dl) pyknotic nuclei (arrow head) of a dying cell; (D2) floating remnants of dead cell organelles including rough ER (arrows).
  • FIG 8 shows result of Pupillary Light Responses of one retina of an adult man with LCA5 (red trace - top line) having similar temporal characteristics as those of an age-comparable normal-sighted man (blue trace - bottom line). The amplitude of constriction was reduced in the LCA5 patient compared to the normal individual.
  • FIG 9 is a table of the thickness of various retinal layers showing that outer nuclear layer (ONL) thickness remains thicker over at least 3 months in the treated (*) compared to the control retinas. There was not such a clear trend in the other retinal layers (outer plexiform layer, OPL; inner nuclear layer, INL, inner plexiform layer,IPL). For each time point, bars from left to right represent thicknesses of ONL, OPL, INL and IPL, respectively.
  • OPL outer nuclear layer
  • INL inner nuclear layer
  • IPL inner plexiform layer
  • FIG 10 provides results of Light-mediated changes in position of phototransduction-specific molecules after injection with AAV7m8.hopt.LCA5.
  • FIGs 1 lA- 1 IB provide a plasmid map and a feature list of the pAAV.CMV.CBA.human native Lebercilin vector.
  • the nucleic acid sequence is reproduced in SEQ ID NO: 9.
  • FIGs 12A- 12F demonstrate cilia phenotype rescued in homozygous human LCA5 p.(Q279*) iPSC-RPE after treatment with AAV7m8.hopt-LCA5.
  • A Confocal images show hexagonal morphology of mature RPE cells along with the immunofluorescently detectable RPE markers; ZO- 1 and MITF.
  • Phase contrast (PC) images display the architecture of iPSC-RPE cultures of RPE derived from both the normal-sighted person and LCA5 patient;
  • Lebercilin is present in normal-sighted control and AAV7m8.LCA5-treated (but not AAV7m8.eGFP-treated) LCA5-affected cells.
  • F Quantitative analysis of number of cilia per cell in normal-sighted- vs. LCA5-iPSC-RPE shows rescue effect of cilia formation after treatment of LCA5-iPSC-RPE cells with AAV7m8.LCA5 (but not with AAV7m8.eGFP).
  • LCA5 nucleic acid sequence encoding Lebercilin protein in one embodiment, such compositions involve codon optimization of Lebercilin coding sequence. It is desirable to increase the efficacy of the product, and thus, increase safety, since a lower dose of reagent may be used.
  • Lebercilin is encoded by the LCA5 gene on chromosome 6ql4 and is a ciliary protein that localizes to the connecting cilia of photoreceptors and to the microtubules, centrioles and primary cilia of cultured mammalian cells.
  • Lebercilin is expressed widely throughout development and is found in cilia of cultured cells as well as in the connecting cilium of mature photoreceptor cells.
  • the connecting cilium is a narrow structure between the photoreceptor inner segment that harbors the biosynthetic machinery of the cell, and the outer segments that contain the opsin-driven visual cascade.
  • the connecting cilium functions as a conduit, supporting the bi-directional trafficking of proteins and vesicles along ciliary microtubule tracks in a process known as intraflagellar transport (IFT).
  • IFT intraflagellar transport
  • Lca5 null mice lack cone and rod ERG responses and undergo an early and progressive retinal degeneration with only a single row of dispersed nuclei (compared to 8- 10 rows of contiguous cells in retinas of wildtype mice) present in the outer nuclear layer (ONL) by 2 months of age l9.
  • LCA5 Leber Congenital Amaurosis
  • the phenotype in affected individuals is limited to the eye and results in blindness.
  • 5 had homozygous nonsense and frameshift mutations and in one family the LCA5 transcript was completely absent.
  • the nucleic acids encoding the Lebercilin cDNA or a codon-optimized version thereof are of the appropriate size to fit into an adeno-associated virus (AAV) vector. See e.g., the sequences in FIGs. 1A and IE to IF and FIG. 1 lA-1 IB.
  • AAV adeno-associated virus
  • retinal degeneration due to LCA5 mutations can be corrected.
  • Such therapy is particularly advantageous if the wildtype or optimized copy of the gene is delivered early in life, e.g., in childhood or in early postnatal period.
  • this intravitreal or subretinal administration used in one embodiment, provides the gene efficiently to the target cells (e.g., photoreceptors).
  • the Lebercilin gene, LCA5, encodes Lebercilin, a 697 amino acid protein thought to be involved in centrosomal or ciliary functions and minus end-directed microtubule transport.
  • LCA5 The Lebercilin gene, LCA5, encodes Lebercilin, a 697 amino acid protein thought to be involved in centrosomal or ciliary functions and minus end-directed microtubule transport.
  • the terms “LCA5" and "Lebercilin” are used interchangeably when referring to the coding sequence.
  • the native nucleic acid sequences encoding human Lebercilin are reported at NCBI Reference Sequence NM_181714.3 (transcript variant 1), NM_001122769.2 (transcript variant 2), XM_01 1535504.1 (transcript variant XI) and XM_005248665.4 (transcript variant X2), and reproduced here in SEQ ID NO: 4, 5, 6 and 7, respectively.
  • Leber congenital amaurosis is an eye disorder that primarily affects the retina, which is the specialized tissue at the back of the eye that detects light and color. People with this disorder typically have severe visual impairment beginning in infancy. The visual impairment tends to be stable, although it may worsen very slowly over time. Leber congenital amaurosis is also associated with other vision problems, including an increased sensitivity to light
  • the pupils which usually expand and contract in response to the amount of light entering the eye, do not react normally to light. Instead, they expand and contract more slowly than normal, or they may not respond to light at all.
  • the clear front covering of the eye may be cone-shaped and abnormally thin, a condition known as keratoconus.
  • a specific behavior called Franceschetti's oculo-digital sign is characteristic of Leber congenital amaurosis. This sign consists of poking, pressing, and rubbing the eyes with a knuckle or finger. researchers suspect that this behavior may contribute to deep-set eyes and keratoconus in affected children.
  • Leber congenital amaurosis In rare cases, delayed development and intellectual disability have been reported in people with the features of Leber congenital amaurosis. However, researchers are uncertain whether these individuals actually have Leber congenital amaurosis or another syndrome with similar signs and symptoms. At least 13 types of Leber congenital amaurosis have been described. The types are distinguished by their genetic cause, patterns of vision loss, and related eye abnormalities.
  • sequence identity refers to the residues in the two sequences which are the same when aligned for correspondence.
  • the length of sequence identity comparison may be over the full-length of the genome, the full-length of a gene coding sequence, or a fragment of at least about 500 to 5000 nucleotides, is desired. However, identity among smaller fragments, e.g. of at least about nine nucleotides, usually at least about 20 to 24 nucleotides, at least about 28 to 32 nucleotides, at least about 36 or more nucleotides, may also be desired.
  • Percent identity may be readily determined for amino acid sequences over the full-length of a protein, polypeptide, about 32 amino acids, about 330 amino acids, or a peptide fragment thereof or the corresponding nucleic acid sequence coding sequences.
  • a suitable amino acid fragment may be at least about 8 amino acids in length, and may be up to about 700 amino acids.
  • identity is determined in reference to "aligned” sequences.
  • Alignments refer to multiple nucleic acid sequences or protein (amino acids) sequences, often containing corrections for missing or additional bases or amino acids as compared to a reference sequence. Identity may be determined by preparing an alignment of the sequences and through the use of a variety of algorithms and/or computer programs known in the art or commercially available [e.g., BLAST, ExPASy; ClustalO; FASTA; using, e.g., Needleman-Wunsch algorithm, Smith-Waterman algorithm] . Alignments are performed using any of a variety of publicly or commercially available Multiple Sequence Alignment Programs.
  • Sequence alignment programs are available for amino acid sequences, e.g., the "Clustal Omega” “Clustal X”, “MAP”, “PIMA”, “MSA”, “BLOCKMAKER”, “MEME”, and “Match-Box” programs. Generally, any of these programs are used at default settings, although one of skill in the art can alter these settings as needed. Alternatively, one of skill in the art can utilize another algorithm or computer program which provides at least the level of identity or alignment as that provided by the referenced algorithms and programs. See, e.g., J. D. Thomson et al, Nucl. Acids. Res., "A comprehensive comparison of multiple sequence alignments", 27(13):2682-2690 (1999).
  • sequence alignment programs are also available for nucleic acid sequences. Examples of such programs include, “Clustal Omega” “Clustal W”, “CAP Sequence Assembly”, “BLAST”, “MAP”, and “MEME”, which are accessible through Web Servers on the internet.
  • nucleotide sequence identity can be measured using FastaTM, a program in GCG Version 6.1.
  • FastaTM provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences. For instance, percent sequence identity between nucleic acid sequences can be determined using FastaTM with its default parameters (a word size of 6 and the NOP AM factor for the scoring matrix) as provided in GCG Version 6.1, herein incorporated by reference.
  • the codon optimized Lebercilin coding sequence has less than about 80% identity, preferably about 75% identity or less to the full-length native Lebercilin coding sequence (FIGs. IB- ID, SEQ ID NO: 2). In one embodiment, the codon optimized Lebercilin coding sequence has about 74% identity with the native Lebercilin coding sequence of SEQ ID NO: 2. In one embodiment, the codon optimized Lebercilin coding sequence is characterized by improved translation rate as compared to native Lebercilin following AAV- mediated delivery (e.g., rAAV).
  • AAV- mediated delivery e.g., rAAV
  • the codon optimized Lebercilin coding sequence shares less than about 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80%, 79%, 78%, 77%, 76%, 75%, 74%, 73%, 72%, 71%, 70%, 69%, 68%, 67%, 66%, 65%, 64%, 63%, 62%, 61% or less identity to the full length native Lebercilin coding sequence of SEQ ID NO: 2.
  • the codon optimized nucleic acid sequence is a variant of SEQ ID NO: 3.
  • the codon optimized nucleic acid sequence a sequence sharing about 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80%, 79%, 78%, 77%, 76%, 75%, 74%, 73%, 72%, 71%, 70%, 69%, 68%, 67%, 66%, 65%, 64%, 63%, 62%, 61% or greater identity with SEQ ID NO: 3.
  • the codon optimized nucleic acid sequence is SEQ ID NO: 3.
  • the nucleic acid sequence is codon optimized for expression in humans.
  • the lebercilin coding sequence is nt 1883 to nt 3976 of SEQ ID NO: 8. In other embodiments, a different Lebercilin coding sequence is selected.
  • Codon-optimized coding regions can be designed by various different methods. This optimization may be performed using methods which are available on-line (e.g., GeneArt), published methods, or a company which provides codon optimizing services, e.g., DNA2.0
  • the entire length of the open reading frame (ORF) for the product is modified.
  • only a fragment of the ORF may be altered.
  • oligonucleotide pairs are synthesized such that upon annealing, they form double stranded fragments of 80-90 base pairs, containing cohesive ends, e.g., each oligonucleotide in the pair is synthesized to extend 3, 4, 5, 6, 7, 8, 9, 10, or more bases beyond the region that is complementary to the other oligonucleotide in the pair.
  • the single-stranded ends of each pair of oligonucleotides are designed to anneal with the single-stranded end of another pair of oligonucleotides.
  • the oligonucleotide pairs are allowed to anneal, and approximately five to six of these double-stranded fragments are then allowed to anneal together via the cohesive single stranded ends, and then they ligated together and cloned into a standard bacterial cloning vector, for example, a TOPO® vector available from Invitrogen Corporation, Carlsbad, Calif.
  • the construct is then sequenced by standard methods. Several of these constructs consisting of 5 to 6 fragments of 80 to 90 base pair fragments ligated together, i.e., fragments of about 500 base pairs, are prepared, such that the entire desired sequence is represented in a series of plasmid constructs.
  • the inserts of these plasmids are then cut with appropriate restriction enzymes and ligated together to form the final construct.
  • the final construct is then cloned into a standard bacterial cloning vector, and sequenced. Additional methods would be immediately apparent to the skilled artisan. In addition, gene synthesis is readily available commercially.
  • the nucleic acid sequences encoding the Lebercilin protein described herein are assembled and placed into any suitable genetic element, e.g., naked DNA, phage, transposon, cosmid, episome, etc., which transfers the Lebercilin sequences carried thereon to a host cell, e.g., for generating non-viral delivery systems (e.g., RNA -based systems, naked DNA, or the like) or for generating viral vectors in a packaging host cell and/or for delivery to a host cells in a subject.
  • the genetic element is a plasmid.
  • engineered constructs are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. See, e.g., Green and Sambrook, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, NY (2012).
  • the term “host cell” may refer to the packaging cell line in which a recombinant AAV is produced from a production plasmid.
  • the term “host cell” may refer to any target cell in which expression of the coding sequence is desired.
  • a “host cell” refers to a prokaryotic or eukaryotic cell that contains exogenous or heterologous DNA that has been introduced into the cell by any means, e.g., electroporation, calcium phosphate precipitation, microinjection, transformation, viral infection, transfection, liposome delivery, membrane fusion techniques, high velocity DNA-coated pellets, viral infection and protoplast fusion.
  • the term “host cell” refers to the cells employed to generate and package the viral vector or recombinant virus.
  • the term “host cell” refers to cultures of ocular cells of various mammalian species for in vitro assessment of the compositions described herein. Still in other embodiments, the term “host cell” is intended to reference the ocular cells of the subject being treated in vivo for LCA.
  • the term "ocular cells" refers to any cell in, or associated with the function of, the eye.
  • the term may refer to any one of photoreceptor cells, including rod photoreceptors, cone photoreceptors and photosensitive ganglion cells, retinal pigment epithelium (RPE) cells, Mueller cells, choroidal cells, bipolar cells, horizontal cells, and amacrine cells.
  • the ocular cells are the photoreceptor cells.
  • the ocular cells are cone photoreceptors.
  • the ocular cells are rod photoreceptors.
  • the nucleic acid sequence encoding Lebercilin further comprises a nucleic acid encoding a tag polypeptide covalently linked thereto.
  • the tag polypeptide may be selected from known "epitope tags" including, without limitation, a myc tag polypeptide, a glutathione-S -transferase tag polypeptide, a green fluorescent protein tag polypeptide, a myc- pyruvate kinase tag polypeptide, a His6 tag polypeptide, an influenza virus hemagglutinin tag polypeptide, a flag tag polypeptide, and a maltose binding protein tag polypeptide.
  • an expression cassette comprising a nucleic acid sequence that encodes
  • Lebercilin is provided.
  • the sequence is a codon optimized sequence.
  • the codon optimized nucleic acid sequence is SEQ ID NO: 3 encoding human Lebercilin.
  • an "expression cassette” refers to a nucleic acid molecule which comprises the coding sequences for Lebercilin protein, promoter, and may include other regulatory sequences therefor, which cassette may be packaged into the capsid of a viral vector (e.g., a viral particle).
  • a viral vector e.g., a viral particle
  • such an expression cassette for generating a viral vector contains the LCA5 sequences described herein flanked by packaging signals of the viral genome and other expression control sequences such as those described herein.
  • the packaging signals are the 5 ' inverted terminal repeat (ITR) and the 3 ' ITR.
  • an expression cassette comprises a codon optimized nucleic acid sequence that encodes Lebercilin protein.
  • the cassette provides the codon optimized LCA5 operatively associated with expression control sequences that direct expression of the codon optimized nucleic acid sequence that encodes Lebercilin in a host cell.
  • the vector genome is the sequence of nt 1-4379 of SEQ ID NO: 8.
  • the vector genome is the sequence of nt 1-4368 of SEQ ID NO: 9.
  • the LCA5 coding sequence in either of the identified vector genomes is swapped with another LCA5 coding sequence as described herein.
  • an expression cassette for use in an AAV vector is provided.
  • the AAV expression cassette includes at least one AAV inverted terminal repeat (ITR) sequence.
  • the expression cassette comprises 5' ITR sequences and 3' ITR sequences.
  • the 5' and 3' ITRs flank the codon optimized nucleic acid sequence that encodes Lebercilin, optionally with additional sequences which direct expression of the codon optimized nucleic acid sequence that encodes Lebercilin in a host cell.
  • a AAV expression cassette is meant to describe an expression cassette as described above flanked on its 5 ' end by a 5 'AAV inverted terminal repeat sequence (ITR) and on its 3 ' end by a 3 ' AAV ITR.
  • ITR inverted terminal repeat sequence
  • this rAAV genome contains the minimal sequences required to package the expression cassette into an AAV viral particle, i.e., the AAV 5 ' and 3 ' ITRs.
  • the AAV ITRs may be obtained from the ITR sequences of any AAV, such as described herein. These ITRs may be of the same AAV origin as the capsid employed in the resulting recombinant AAV, or of a different AAV origin (to produce an AAV pseudotype).
  • the ITR sequences from AAV2, or the deleted version thereof (AITR) are used for convenience and to accelerate regulatory approval.
  • ITRs from other AAV sources may be selected.
  • the source of the ITRs is from AAV2 and the AAV capsid is from another AAV source
  • the resulting vector may be termed pseudotyped.
  • the AAV vector genome comprises an AAV 5 ' ITR, the Lebercilin coding sequences and any regulatory sequences, and an AAV 3 ' ITR.
  • AITR A shortened version of the 5 ' ITR, termed AITR, has been described in which the D- sequence and terminal resolution site (trs) are deleted.
  • trs D- sequence and terminal resolution site
  • the full-length AAV 5' and 3' ITRs are used.
  • Each rAAV genome can be then introduced into a production plasmid.
  • regulatory sequences refers to DNA sequences, such as initiator sequences, enhancer sequences, and promoter sequences, which induce, repress, or otherwise control the transcription of protein encoding nucleic acid sequences to which they are operably linked.
  • operably linked refers to both expression control sequences that are contiguous with the nucleic acid sequence encoding the Lebercilin and/or expression control sequences that act in trans or at a distance to control the transcription and expression thereof.
  • a vector comprising any of the expression cassettes described herein is provided. As described herein, such vectors can be plasmids of variety of origins and are useful in certain embodiments for the generation of recombinant replication defective viruses as described further herein.
  • a "vector” as used herein is a nucleic acid molecule into which an exogenous or heterologous or engineered nucleic acid transgene may be inserted which can then be introduced into an appropriate host cell.
  • Vectors preferably have one or more origin of replication, and one or more site into which the recombinant DNA can be inserted.
  • Vectors often have means by which cells with vectors can be selected from those without, e.g., they encode drug resistance genes.
  • Common vectors include plasmids, viral genomes, and (primarily in yeast and bacteria) "artificial chromosomes.” Certain plasmids are described herein.
  • the vector is a non-viral plasmid that comprises an expression cassette described thereof, e.g., "naked DNA”, “naked plasmid DNA”, RNA, and mRNA;
  • compositions and nano particles including, e.g., micelles, liposomes, cationic lipid - nucleic acid compositions, poly-glycan compositions and other polymers, lipid and/or cholesterol-based - nucleic acid conjugates, and other constructs such as are described herein. See, e.g., X. Su et al, Mol. Pharmaceutics, 201 1, 8 (3), pp 774-787; web publication: March 21, 201 1 ; WO2013/182683, WO 2010/053572 and WO 2012/170930, all of which are incorporated herein by reference.
  • Such non-viral Lebercilin vector may be administered by the routes described herein.
  • the viral vectors, or non-viral vectors can be formulated with a physiologically acceptable carrier for use in gene transfer and gene therapy applications.
  • the vector is a viral vector that comprises an expression cassette described therein.
  • Virus vectors are defined as replication defective viruses containing the exogenous or heterologous LCA5 nucleic acid transgene.
  • an expression cassette as described herein may be engineered onto a plasmid which is used for drug delivery or for production of a viral vector.
  • Suitable viral vectors are preferably replication defective and selected from amongst those which target ocular cells.
  • Viral vectors may include any virus suitable for gene therapy, including but not limited to adenovirus; herpes virus; lentivirus;
  • adeno-associated virus is referenced herein as an exemplary virus vector.
  • a “replication-defective virus” or “viral vector” refers to a synthetic or recombinant viral particle in which an expression cassette containing a gene of interest is packaged in a viral capsid or envelope, where any viral genomic sequences also packaged within the viral capsid or envelope are replication- deficient; i.e., they cannot generate progeny virions but retain the ability to infect target cells.
  • the genome of the viral vector does not include genes encoding the enzymes required to replicate (the genome can be engineered to be "gutless" - containing only the transgene of interest flanked by the signals required for amplification and packaging of the artificial genome), but these genes may be supplied during production.
  • a recombinant adeno-associated virus (rAAV) vector is provided.
  • the rAAV compromises an AAV capsid, and a vector genome packaged therein.
  • the vector genome comprises, in one embodiment: (a) an AAV 5' inverted terminal repeat (ITR) sequence; (b) a promoter; (c) a coding sequence encoding a human Lebercilin; and (d) an AAV 3' ITR.
  • the vector genome is the expression cassette described herein.
  • the LCA5 sequence encodes a full length Lebercilin protein.
  • the Lebercilin sequence is the protein sequence of SEQ ID NO: 1.
  • the coding sequence is SEQ ID NO: 3 or a variant thereof.
  • Adeno-associated virus a member of the Parvovirus family, is a small nonenveloped, icosahedral virus with single-stranded linear DNA genomes of 4.7 kilobases (kb) to 6 kb.
  • AAV serotypes are AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9 and others.
  • the ITRs or other AAV components may be readily isolated or engineered using techniques available to those of skill in the art from an AAV. Such AAV may be isolated, engineered, or obtained from academic, commercial, or public sources (e.g., the American Type Culture Collection, Manassas, VA).
  • the AAV sequences may be engineered through synthetic or other suitable means by reference to published sequences such as are available in the literature or in databases such as, e.g., GenBank, PubMed, or the like.
  • AAV viruses may be engineered by conventional molecular biology techniques, making it possible to optimize these particles for cell specific delivery of nucleic acid sequences, for minimizing immunogenicity, for tuning stability and particle lifetime, for efficient degradation, for accurate delivery to the nucleus, etc.
  • Fragments of AAV may be readily utilized in a variety of vector systems and host cells.
  • AAV fragments include the cap proteins, including the vp l, vp2, vp3 and hypervariable regions, the rep proteins, including rep 78, rep 68, rep 52, and rep 40, and the sequences encoding these proteins.
  • Such fragments may be used alone, in combination with other AAV serotype sequences or fragments, or in combination with elements from other AAV or non-AAV viral sequences.
  • artificial AAV serotypes include, without limitation, AAV with a non-naturally occurring capsid protein.
  • Such an artificial capsid may be generated by any suitable technique, using a novel AAV sequence of the invention (e.g., a fragment of a vpl capsid protein) in combination with heterologous sequences which may be obtained from another AAV serotype (known or novel), non-contiguous portions of the same AAV serotype, from a non-AAV viral source, or from a non-viral source.
  • An artificial AAV serotype may be, without limitation, a chimeric AAV capsid, a recombinant AAV capsid, or a "humanized" AAV capsid.
  • a vector contains the AAV8 cap and/or rep sequences of the invention. See e.g., US patent application publication No. US2009/02270030, incorporated by reference herein.
  • AAV or "AAV serotype” as used herein refers to the dozens of naturally occurring and available adeno-associated viruses, as well as artificial AAVs.
  • human AAV2 is the first AAV that was developed as a gene transfer vector; it has been widely used for efficient gene transfer experiments in different target tissues and animal models.
  • the AAV capsid, ITRs, and other selected AAV components described herein may be readily selected from among any AAV, including, without limitation, AAV 1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV8bp, AAV7M8 and
  • the AAV capsid is an AAV8bp capsid, which preferentially targets bipolar cells. See, WO 2014/024282, which is incorporated herein by reference.
  • the AAV capsid is an AAV7m8 capsid, which has shown preferential delivery to the outer retina.
  • the AAV7m8 capsid nucleic acid sequence is reproduced in SEQ ID NO: 11 and amino acid sequence at SEQ ID NO: 12.
  • an "AAV7m8 capsid” is a self-assembled AAV capsid composed of multiple AAV7m8 vp (variable protein) proteins.
  • the AAV7m8 vp proteins are typically expressed as alternative splice variants encoded by a nucleic acid sequence of SEQ ID NO: 1 1 or a sequence at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 99% thereto, which encodes the vp l amino acid sequence of SEQ ID NO: 12.
  • These splice variants result in proteins of different length of SEQ ID NO: 12.
  • "AAV7m8 capsid” includes an AAV having an amino acid sequence which is 99% identical to SEQ ID NO: 12.
  • the rAAV capsid is selected from an AAV8 capsid or variant thereof, an AAV6 capsid or variant thereof, an AAV9 capsid or variant thereof, an AAV7 capsid or variant thereof, an AAV5 capsid or variant thereof, an AAV2 capsid or variant thereof, an AAV1 capsid or variant thereof, an AAV3 capsid or variant thereof, and an AAV4 capsid or variant thereof.
  • a recombinant adeno-associated virus (rAAV) vector which comprises an AAV7m8 capsid and an expression cassette described herein, wherein said expression cassette comprises nucleic acid sequences encoding Lebercilin, inverted terminal repeat sequences and expression control sequences that direct expression of Lebercilin in a host cell.
  • rAAV adeno-associated virus
  • a recombinant adeno-associated virus (AAV) vector is provided for delivery of the LCA5 constructs and optimized sequences described herein.
  • An adeno-associated virus (AAV) viral vector is an AAV DNase-resistant particle having an AAV protein capsid into which is packaged nucleic acid sequences for delivery to target cells.
  • An AAV capsid is composed of 60 capsid (cap) protein subunits, VP 1, VP2, and VP3, that are arranged in an icosahedral symmetry in a ratio of approximately 1 : 1 : 10 to 1 : 1 :20, depending upon the selected AAV.
  • AAVs may be selected as sources for capsids of AAV viral vectors as identified above. See, e.g., US Published Patent Application No. 2007-0036760-A1 ; US
  • an AAV cap for use in the viral vector can be generated by mutagenesis (i.e., by insertions, deletions, or substitutions) of one of the aforementioned AAV capsids or its encoding nucleic acid.
  • the AAV capsid is chimeric, comprising domains from two or three or four or more of the aforementioned AAV capsid proteins.
  • the AAV capsid is a mosaic of Vpl, Vp2, and Vp3 monomers from two or three different AAVs or recombinant AAVs.
  • an rAAV composition comprises more than one of the aforementioned Caps.
  • the term variant means any AAV sequence which is derived from a known AAV sequence, including those sharing at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 99% or greater sequence identity over the amino acid or nucleic acid sequence.
  • the AAV capsid includes variants which may include up to about 10% variation from any described or known AAV capsid sequence. That is, the AAV capsid shares about 90% identity to about 99.9 % identity, about 95% to about 99% identity or about 97% to about 98% identity to an AAV capsid provided herein and/or known in the art.
  • the AAV capsid shares at least 95% identity with an AAV capsid.
  • the comparison may be made over any of the variable proteins (e.g., vpl, vp2, or vp3).
  • the AAV capsid shares at least 95% identity with the AAV7m8 over the vp l, vp2 or vp3.
  • the capsid is an AAV8 capsid with Y447F, Y733F and T494V mutations (also called “AAV8(C&G+T494V)” and M rep2-cap8(Y447F+733F+T494V)”), as described by Kay et al, Targeting Photoreceptors via Intravitreal Delivery Using Novel, Capsid- Mutated AAV Vectors, PLoS One. 2013; 8(4): e62097. Published online 2013 Apr 26, which is incorporated herein by reference.
  • an AAV capsid which shows tropism for the desired target cell, e.g., photoreceptors (e.g., rods and/or cones), RPE or other ocular cells.
  • the AAV capsid is a tyrosine capsid-mutant in which certain surface exposed tyrosine residues are substituted with phenylalanine (F).
  • F phenylalanine
  • artificial AAV means, without limitation, an AAV with a non-naturally occurring capsid protein.
  • Such an artificial capsid may be generated by any suitable technique, using a selected AAV sequence (e.g., a fragment of a vpl capsid protein) in combination with heterologous sequences which may be obtained from a different selected AAV, non-contiguous portions of the same AAV, from a non-AAV viral source, or from a non-viral source.
  • An artificial AAV may be, without limitation, a pseudotyped AAV, a chimeric AAV capsid, a recombinant AAV capsid, or a "humanized" AAV capsid.
  • Pseudotyped vectors, wherein the capsid of one AAV is replaced with a heterologous capsid protein, are useful in the invention.
  • AAV2/5 and AAV2/8 are exemplary pseudotyped vectors.
  • a self-complementary AAV refers a plasmid or vector having an expression cassette in which a coding region carried by a recombinant AAV nucleic acid sequence has been designed to form an intra-molecular double- stranded DNA template.
  • dsDNA double stranded DNA
  • exogenous nucleic acid sequence or protein means that the nucleic acid or protein does not naturally occur in the position in which it exists in a chromosome, or host cell.
  • An exogenous nucleic acid sequence also refers to a sequence derived from and inserted into the same host cell or subject, but which is present in a non-natural state, e.g. a different copy number, or under the control of different regulatory elements.
  • heterologous as used to describe a nucleic acid sequence or protein means that the nucleic acid or protein was derived from a different organism or a different species of the same organism than the host cell or subject in which it is expressed.
  • heterologous when used with reference to a protein or a nucleic acid in a plasmid, expression cassette, or vector, indicates that the protein or the nucleic acid is present with another sequence or subsequence which with which the protein or nucleic acid in question is not found in the same relationship to each other in nature.
  • the expression cassette including any of those described herein is employed to generate a recombinant AAV genome.
  • the expression cassette described herein is engineered into a suitable genetic element (vector) useful for generating viral vectors and/or for delivery to a host cell, e.g., naked DNA, phage, transposon, cosmid, episome, etc., which transfers the LCA5 sequences carried thereon.
  • a suitable genetic element useful for generating viral vectors and/or for delivery to a host cell, e.g., naked DNA, phage, transposon, cosmid, episome, etc., which transfers the LCA5 sequences carried thereon.
  • the selected vector may be delivered by any suitable method, including transfection, electroporation, liposome delivery, membrane fusion techniques, high velocity DNA-coated pellets, viral infection and protoplast fusion.
  • the methods used to make such constructs are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. See, e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring
  • the ITRs are the only AAV components required in cis in the same construct as the expression cassette.
  • the coding sequences for the replication (rep) and/or capsid (cap) are removed from the AAV genome and supplied in trans or by a packaging cell line in order to generate the AAV vector.
  • a producer cell line is transiently transfected with a construct that encodes the transgene flanked by ITRs and a construct(s) that encodes rep and cap.
  • a packaging cell line that stably supplies rep and cap is transiently transfected with a construct encoding the transgene flanked by ITRs.
  • AAV virions are produced in response to infection with helper adenovirus or herpesvirus, requiring the separation of the rAAVs from contaminating virus.
  • helper adenovirus or herpesvirus More recently, systems have been developed that do not require infection with helper virus to recover the AAV - the required helper functions (i.e., adenovirus El, E2a, VA, and E4 or herpesvirus UL5, UL8, UL52, and UL29, and herpesvirus polymerase) are also supplied, in trans, by the system.
  • helper functions can be supplied by transient transfection of the cells with constructs that encode the required helper functions, or the cells can be engineered to stably contain genes encoding the helper functions, the expression of which can be controlled at the transcriptional or posttranscriptional level.
  • isolated means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally -occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same
  • polynucleotide or polypeptide separated from some or all of the coexisting materials in the natural system, is isolated, even if subsequently reintroduced into the natural system.
  • Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
  • the expression cassette flanked by ITRs and rep/cap genes are introduced into insect cells by infection with baculovirus-based vectors.
  • baculovirus-based vectors For reviews on these production systems, see generally, e.g., Zhang et al, 2009, "Adenovirus-adeno-associated virus hybrid for large-scale recombinant adeno-associated virus production," Human Gene Therapy 20:922-929, the contents of which is incorporated herein by reference in its entirety. Methods of making and using these and other AAV production systems are also described in the following U.S.
  • any embodiment of this invention is known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. See, e.g., Green and Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, NY (2012). Similarly, methods of generating rAAV virions are well known and the selection of a suitable method is not a limitation on the present invention. See, e.g., K. Fisher et al, (1993) J. Virol, 70:520-532 and US Patent No. 5,478,745.
  • Plasmids generally are designated herein by a lower case p preceded and/or followed by capital letters and/or numbers, in accordance with standard naming conventions that are familiar to those of skill in the art. Many plasmids and other cloning and expression vectors that can be used in accordance with the present invention are well known and readily available to those of skill in the art. Moreover, those of skill readily may construct any number of other plasmids suitable for use in the invention. The properties, construction and use of such plasmids, as well as other vectors, in the present invention will be readily apparent to those of skill from the present disclosure.
  • the production plasmid is that described herein, or as described in WO2012/158757, which is incorporated herein by reference.
  • Various plasmids are known in the art for use in producing rAAV vectors, and are useful herein.
  • the production plasmids are cultured in the host cells which express the AAV cap and/or rep proteins. In the host cells, each rAAV genome is rescued and packaged into the capsid protein or envelope protein to form an infectious viral particle.
  • a production plasmid comprising an expression cassette described above is provided.
  • the production plasmid is that shown in SEQ ID NO: 8, and FIG. IE- IF, which is termed p643.
  • This plasmid is used in the examples for generation of the rAAV- human codon optimized Lebercilin vector.
  • a plasmid is one that contains a 5' AAV ITR sequence; a selected promoter; a polyA sequence; and a 3' ITR; additionally, it also contains a staffer sequence, such as lambda.
  • the staffer sequence keeps the rAAV vector genome with a size between about 3 kilobases (kb) to about 6 kb, about 4.7 kb to about 6 kb, about 3 kb to about 5.5kb, or about 4.7 kb to 5.5 kb.
  • a non-coding lambda staffer region is included in the vector backbone.
  • the production plasmid is that shown in FIG. 1 lA- 1 IB and SEQ ID NO: 9.
  • the production plasmid is modified to optimized vector plasmid production efficiency. Such modifications include addition of other neutral sequences, or deletion of portion(s) of or the entire lambda staffer sequence to modulate the level of supercoil of the vector plasmid. Such modifications are contemplated herein. In other embodiments, terminator and other sequences are included in the plasmid.
  • the rAAV expression cassette, the vector (such as rAAV vector), the virus (such as rAAV), the production plasmid comprises AAV inverted terminal repeat sequences, a codon optimized nucleic acid sequence that encodes Lebercilin, and expression control sequences that direct expression of the encoded proteins in a host cell.
  • the rAAV expression cassette, the virus, the vector (such as rAAV vector), the production plasmid further comprise one or more of an intron, a Kozak sequence, a polyA, posttranscriptional regulatory elements and others.
  • the post-transcriptional regulatory element is Woodchuck Hepatitis Virus (WHP) Posttranscriptional Regulatory Element (WPRE).
  • the expression cassettes, vectors and plasmids include other components that can be optimized for a specific species using techniques known in the art including, e.g, codon optimization, as described herein.
  • the components of the cassettes, vectors, plasmids and viruses or other compositions described herein include a promoter sequence as part of the expression control sequences.
  • the promoter is cell-specific.
  • the term "cell-specific" means that the particular promoter selected for the recombinant vector can direct expression of the optimized Lebercilin coding sequence in a particular ocular cell type.
  • the promoter is specific for expression of the transgene in photoreceptor cells.
  • the promoter is specific for expression in the rods and cones.
  • the promoter is specific for expression in the rods. In another embodiment, the promoter is specific for expression in the cones. In one embodiment, the photoreceptor- specific promoter is a human rhodopsin kinase promoter. The rhodopsin kinase promoter has been shown to be active in both rods and cones. See, e.g., Sun et al, Gene Therapy with a Promoter Targeting Both Rods and Cones Rescues Retinal Degeneration Caused by AIPL 1 Mutations, Gene Ther. 2010 January; 17(1): 117-131, which is incorporated herein by reference in its entirety. In one embodiment, the promoter is modified to add one or more restriction sites to facilitate cloning.
  • the promoter is a human rhodopsin promoter.
  • the promoter is modified to include restriction on the ends for cloning. See, e.g, Nathans and Hogness, Isolation and nucleotide sequence of the gene encoding human rhodopsin, PNAS, 81 :4851-5 (August 1984), which is incorporated herein by reference in its entirety.
  • the promoter is a portion or fragment of the human rhodopsin promoter.
  • the promoter is a variant of the human rhodopsin promoter.
  • exemplary promoters include the human G-protein-coupled receptor protein kinase
  • the promoter is a 292 nt fragment (positions 1793-2087) of the GRKl promoter (See, Beltran et al, Gene Therapy 2010 17: 1162-74, which is hereby incorporated by reference in its entirety).
  • the promoter is the human interphotoreceptor retinoid-binding protein proximal (IRBP) promoter.
  • the promoter is a 235 nt fragment of the hIRBP promoter.
  • the promoter is the RPGR proximal promoter (Shu et al, IOVS, May 2102, which is incorporated by reference in its entirety).
  • promoters useful in the invention include, without limitation, the rod opsin promoter, the red-green opsin promoter, the blue opsin promoter, the cGMP-P-phosphodiesterase promoter (Qgueta et al, IOVS, Invest Ophthalmol Vis Sci.
  • mice opsin promoter Beltran et al 2010 cited above
  • the rhodopsin promoter Mosolino et al, Gene Ther, July 2011, 18(7):637-45
  • the alpha-subunit of cone transducin Morrissey et al, BMC Dev, Biol, Jan 2011, 1 1 :3)
  • beta phosphodiesterase (PDE) promoter the retinitis pigmentosa (RP l) promoter (Nicord et al, J. Gene Med, Dec 2007, 9(12): 1015-23)
  • the NXNL2/NXNL 1 promoter Libard et al, PLoS One, Oct.
  • the promoter is selected from human human EF la promoter, rhodopsin promoter, rhodopsin kinase, interphotoreceptor binding protein (IRBP), cone opsin promoters (red-green, blue), cone opsin upstream sequences containing the red-green cone locus control region, cone transducing, and transcription factor promoters (neural retina leucine zipper (Nrl) and photoreceptor-specific nuclear receptor Nr2e3, bZIP).
  • human human EF la promoter rhodopsin promoter, rhodopsin kinase
  • IRBP interphotoreceptor binding protein
  • cone opsin promoters red-green, blue
  • cone opsin upstream sequences containing the red-green cone locus control region cone transducing
  • transcription factor promoters neural retina leucine zipper (Nrl) and photoreceptor-specific nuclear receptor Nr2e3, bZIP.
  • the promoter is a ubiquitous or constitutive promoter.
  • An example of a suitable promoter is a hybrid chicken ⁇ -actin (CBA) promoter with cytomegalovirus (CMV) enhancer elements, such as the sequence shown in FIG. IE- IF.
  • the promoter is the CB7 promoter.
  • CBA chicken ⁇ -actin
  • CMV cytomegalovirus
  • Other suitable promoters include the human ⁇ -actin promoter, the human elongation factor- la promoter, the cytomegalovirus (CMV) promoter, the simian virus 40 promoter, and the herpes simplex virus thymidine kinase promoter.
  • promoters include viral promoters, constitutive promoters, regulatable promoters [see, e.g., WO 2011/126808 and WO 2013/04943].
  • a promoter responsive to physiologic cues may be utilized in the expression cassette, rAAV genomes, vectors, plasmids and viruses described herein.
  • the promoter is of a small size, under 1000 bp, due to the size limitations of the AAV vector.
  • the promoter is under 400 bp.
  • Other promoters may be selected by one of skill in the art.
  • the promoter is selected from SV40 promoter, the dihydrofolate reductase promoter, and the phosphoglycerol kinase (PGK) promoter, rhodopsin kinase promoter, the rod opsin promoter, the red-green opsin promoter, the blue opsin promoter, the inter photoreceptor binding protein (IRBP) promoter and the cGMP- -phosphodiesterase promoter, a phage lambda (PL) promoter, a herpes simplex viral (HSV) promoter, a tetracycline-controlled trans-activator-responsive promoter (tet) system, a long terminal repeat (LTR) promoter, such as a RSV LTR, MoMLV LTR, BIV LTR or an HIV LTR, a U3 region promoter of Moloney murine sarcoma virus, a Granzyme A promoter, a regulatory sequence(s) of the metallo
  • the promoter is an inducible promoter.
  • the inducible promoter may be selected from known promoters including the rapamycin/rapalog promoter, the ecdysone promoter, the estrogen-responsive promoter, and the tetracycline-responsive promoter, or heterodimeric repressor switch. See, Sochor et al, An Autogenously Regulated Expression System for Gene Therapeutic Ocular Applications. Scientific Reports, 2015 Nov 24;5 : 17105 and Daber R, Lewis M., A novel molecular switch. J Mol Biol. 2009 Aug 28;391(4):661-70, Epub 2009 Jun 21 which are both incorporated herein by reference in their entirety.
  • the promoter is a chicken beta-actin promoter with a nucleic acid sequence from nt 546 to nt 283 of SEQ ID NO. 8.
  • the expression cassette, vector, plasmid and virus described herein contain other appropriate transcription initiation, termination, enhancer sequences, efficient RNA processing signals such as splicing and polyadenylation (poly A) signals; TATA sequences; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i. e., Kozak consensus sequence); introns; sequences that enhance protein stability; and when desired, sequences that enhance secretion of the encoded product.
  • the expression cassette or vector may contain none, one or more of any of the elements described herein.
  • polyA sequences include, e.g., a synthetic polyA or from bovine growth hormone (bGH), human growth hormone (hGH), SV40, rabbit ⁇ -globin (RGB), or modified RGB (mRGB).
  • bGH bovine growth hormone
  • hGH human growth hormone
  • SV40 rabbit ⁇ -globin
  • RGB rabbit ⁇ -globin
  • mRGB modified RGB
  • the poly A has a nucleic acid sequence from nt 3993 to nt 4200 of SEQ ID NO: 8.
  • Suitable enhancers include, e.g., the CMV enhancer, the RSV enhancer, the alpha fetoprotein enhancer, the TTR minimal promoter/enhancer, LSP (TH -binding globulin promoter/alpha 1-microglobulin/bikunin enhancer), an APB enhancer, ABPS enhancer, an alpha mic/bik enhancer, TTR enhancer, en34, ApoE amongst others.
  • the enhancer has a nucleic acid sequence from nt 241 to nt 544 of SEQ ID NO: 8.
  • a Kozak sequence is included upstream of the Lebercilin coding sequence to enhance translation from the correct initiation codon.
  • CBA exon 1 and intron are included in the expression cassette.
  • the Lebercilin coding sequence is placed under the control of a hybrid chicken ⁇ actin (CBA) promoter. This promoter consists of the cytomegalovirus (CMV) immediate early enhancer, the proximal chicken ⁇ actin promoter, and CBA exon 1 flanked by intron 1 sequences.
  • the intron is selected from CBA, human beta globin, IVS2, SV40, bGH, alpha-globulin, beta-globulin, collagen, ovalbumin, p53, or a fragment thereof.
  • the expression cassette, the vector, the plasmid and the virus contain a 5 ' ITR, chicken beta-actin (CBA) promoter, CMV enhancer, CBA exon 1 and intron, human codon optimized Lebercilin sequence, bGH poly A and 3' ITR.
  • the expression cassette includes nt 1 to 4379 of SEQ ID NO: 8.
  • the 5 ' ITR has a nucleic acid sequence from nt 1 to nt 130 of SEQ ID NO: 8 and the 3 'ITR has a nucleic acid sequence from nt 4250 to nt 4379 of SEQ ID NO: 8.
  • the CBA exonl and intron has a nucleic acid sequence from nt 824 to nt 1795 of SEQ ID NO: 8.
  • the production plasmid has a sequence of SEQ ID NO: 8, also shown in
  • FIGs. IE- IF the production plasmid has a sequence of SEQ ID NO: 9, also shown in FIGs. 1A- 11B.
  • a method for treating Leber Congenital Amaurosis caused by a defect in the lebercilin gene and/or restoring visual function in a subject having LCA comprises delivering to a subject in need thereof a vector (such as rAAV) which encodes Lebercilin, as described herein.
  • a method of treating a subject having LCA with a rAAV described herein is provided.
  • administering means delivering the composition to the target selected cell which is characterized by LCA.
  • the method involves delivering the composition by subretinal injection to the RPE, photoreceptor cells or other ocular cells.
  • intravitreal injection to the subject is employed.
  • subretinal injection to the subject is employed.
  • intravascular injections such as injection via the palpebral vein may be employed. Still other methods of administration may be selected by one of skill in the art given this disclosure.
  • administering or “route of administration” is delivery of composition described herein, with or without a pharmaceutical carrier or excipient, of the subject. Routes of administration may be combined, if desired. In some embodiments, the administration is repeated periodically.
  • the pharmaceutical compositions described herein are designed for delivery to subjects in need thereof by any suitable route or a combination of different routes. In some embodiments, direct delivery to the eye (optionally via ocular delivery, subretinal injection, intra-retinal injection, intravitreal, topical), or delivery via systemic routes is employed, e.g., intravascular, intraarterial, intraocular, intravenous, intramuscular, subcutaneous, intradermal, and other parental routes of administration.
  • the nucleic acid molecules, the expression cassette and/or vectors described herein may be delivered in a single composition or multiple
  • compositions comprising two or more different AAV may be delivered, or multiple viruses [see, e.g., WO20 201 1/126808 and WO 2013/049493].
  • multiple viruses may contain different replication-defective viruses (e.g., AAV and adenovirus), alone or in combination with proteins.
  • compositions are designed for delivery to subjects in need thereof by any suitable route or a combination of different routes. These delivery means are designed to avoid direct systemic delivery of the suspension containing the AAV composition(s) described herein. Suitably, this may have the benefit of reducing dose as compared to systemic administration, reducing toxicity and/or reducing undesirable immune responses to the AAV and/or transgene product.
  • nucleic acid sequences, vectors, expression cassettes and rAAV viral vectors are useful in a pharmaceutical composition, which also comprises a
  • compositions are used to express the optimized Lebercilin in the host cells through delivery by such recombinantly engineered AAVs or artificial AAVs.
  • the sequences or vectors or viral vector is preferably assessed for contamination by conventional methods and then formulated into a pharmaceutical composition suitable for administration to the eye.
  • a pharmaceutically and/or physiologically acceptable vehicle or carrier particularly one suitable for administration to the eye, such as buffered saline or other buffers, e.g., HEPES, to maintain pH at appropriate physiological levels, and, optionally, other medicinal agents, pharmaceutical agents, stabilizing agents, buffers, carriers, adjuvants, diluents, surfactant, or excipient etc.
  • the carrier will typically be a liquid.
  • Exemplary physiologically acceptable carriers include sterile, pyrogen-free water and sterile, pyrogen-free, phosphate buffered saline.
  • the carrier is an isotonic sodium chloride solution.
  • the carrier is balanced salt solution.
  • the carrier includes tween. If the virus is to be stored long-term, it may be frozen in the presence of glycerol or Tween20.
  • the rAAV for administration to a human patient, is suitably suspended in an aqueous solution containing saline, a surfactant, and a physiologically compatible salt or mixture of salts.
  • the formulation is adjusted to a physiologically acceptable pH, e.g., in the range of pH 6 to 9, or pH 6.5 to 7.5, pH 7.0 to 7.7, or pH 7.2 to 7.8.
  • a physiologically acceptable pH e.g., in the range of pH 6 to 9, or pH 6.5 to 7.5, pH 7.0 to 7.7, or pH 7.2 to 7.8.
  • a pH within this range may be desired; whereas for intravitreal or subretinal delivery, a pH of 6.8 to about 7.2 may be desired.
  • other pHs within the broadest ranges and these subranges may be selected for other route of delivery.
  • a suitable surfactant, or combination of surfactants may be selected from among non- ionic surfactants that are nontoxic.
  • a difunctional block copolymer surfactant terminating in primary hydroxyl groups is selected, e.g., such as Pluronic® F68
  • Poloxamer 188 which has a neutral pH, has an average molecular weight of 8400.
  • Other surfactants and other Poloxamers may be selected, i.e., nonionic triblock copolymers composed of a central hydrophobic chain of polyoxypropylene (poly(propylene oxide)) flanked by two hydrophilic chains of poly oxy ethylene (poly(ethylene oxide)), SOLUTOL HS 15 (Macrogol- 15 Hydroxystearate), LABRASOL (Poly oxy capryllic glyceride), poly oxy 10 oleyl ether, TWEEN (polyoxyethylene sorbitan fatty acid esters), ethanol and polyethylene glycol.
  • the formulation contains a poloxamer.
  • poloxamer These copolymers are commonly named with the letter "P" (for poloxamer) followed by three digits: the first two digits x 100 give the approximate molecular mass of the polyoxypropylene core, and the last digit x 10 gives the percentage polyoxyethylene content.
  • Poloxamer 188 is selected.
  • the surfactant may be present in an amount up to about 0.0005 % to about 0.001% of the suspension.
  • the formulation may contain, e.g., buffered saline solution comprising one or more of sodium chloride, sodium bicarbonate, dextrose, magnesium sulfate (e.g., magnesium sulfate -7H20), potassium chloride, calcium chloride (e.g., calcium chloride -2H20), dibasic sodium phosphate, and mixtures thereof, in water.
  • the osmolarity is within a range compatible with cerebrospinal fluid (e.g., about 275 to about 290); see, e.g., emedicine.medscape.com/article/2093316-overview.
  • a commercially available diluent may be used as a suspending agent, or in combination with another suspending agent and other optional excipients. See, e.g., Elliotts B® solution [Lukare Medical] .
  • the formulation may contain one or more permeation enhancers.
  • suitable permeation enhancers may include, e.g., mannitol, sodium glycocholate, sodium taurocholate, sodium deoxycholate, sodium salicylate, sodium caprylate, sodium caprate, sodium lauryl sulfate, polyoxyethylene-9-laurel ether, or EDTA.
  • the composition includes a carrier, diluent, excipient and/or adjuvant.
  • Suitable carriers may be readily selected by one of skill in the art in view of the indication for which the transfer virus is directed.
  • one suitable carrier includes saline, which may be formulated with a variety of buffering solutions (e.g., phosphate buffered saline).
  • Other exemplary carriers include sterile saline, lactose, sucrose, calcium phosphate, gelatin, dextran, agar, pectin, peanut oil, sesame oil, and water.
  • the buffer/carrier should include a component that prevents the rAAV, from sticking to the infusion tubing but does not interfere with the rAAV binding activity in vivo.
  • compositions of the invention may contain, in addition to the rAAV and carrier(s), other conventional pharmaceutical ingredients, such as preservatives, or chemical stabilizers.
  • suitable exemplary preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, and
  • Suitable chemical stabilizers include gelatin and albumin.
  • compositions according to the present invention may comprise a pharmaceutically acceptable carrier, such as defined above.
  • the compositions described herein comprise an effective amount of one or more AAV suspended in a pharmaceutically suitable carrier and/or admixed with suitable excipients designed for delivery to the subject via injection, osmotic pump, intrathecal catheter, or for delivery by another device or route.
  • the composition is formulated for intravitreal delivery.
  • the composition is formulated for subretinal delivery.
  • the composition of the carrier or excipient contains 180 mM NaCl, 10 mM NaPi, pH7.3 with 0.0001% - 0.01% Pluronic F68 (PF68).
  • the exact composition of the saline component of the buffer ranges from 160 mM to 180 mM NaCl.
  • a different pH buffer pH buffer (potentially HEPES, sodium bicarbonate, TRIS) is used in place of the buffer specifically described.
  • a buffer containing 0.9% NaCl is useful.
  • GC genome copies
  • VG vector genomes
  • virus particles may be used as the measure of the dose contained in the formulation or suspension. Any method known in the art can be used to determine the genome copy (GC) number of the replication-defective virus compositions of the invention.
  • One method for performing AAV GC number titration is as follows: Purified AAV vector samples are first treated with DNase to eliminate un-encapsidated AAV genome DNA or contaminating plasmid DNA from the production process. The DNase resistant particles are then subjected to heat treatment to release the genome from the capsid. The released genomes are then quantitated by real-time PCR using primer/probe sets targeting specific region of the viral genome (usually poly A signal).
  • the effective dose of a recombinant adeno-associated virus carrying a nucleic acid sequence encoding the optimized Lebercilin coding sequence is measured as described in S.K. McLaughlin et al, 1988 J. Virol, 62: 1963, which is incorporated by reference in its entirety.
  • the term "dosage” can refer to the total dosage delivered to the subject in the course of treatment, or the amount delivered in a single unit (or multiple unit or split dosage) administration.
  • the pharmaceutical virus compositions can be formulated in dosage units to contain an amount of replication-defective virus carrying the codon optimized nucleic acid sequences encoding Lebercilin as described herein that is in the range of about 1.0 x 10 9 GC to about 1.0 x 10 15 GC per dose including all integers or fractional amounts within the range.
  • the compositions are formulated to contain at least lxlO 9 , 2xl0 9 , 3xl0 9 , 4xl0 9 , 5xl0 9 , 6xl0 9 , 7xl0 9 , 8xl0 9 , or 9xl0 9 GC per dose including all integers or fractional amounts within the range.
  • the compositions are formulated to contain at least lxlO 10 , 2xl0 10 , 3xl0 10 , 4xl0 10 , 5xl0 10 , 6xl0 10 , 7xl0 10 , 8xl0 10 , or 9xl0 10 GC per dose including all integers or fractional amounts within the range.
  • compositions are formulated to contain at least lxlO 11 , 2xlO u , 3xl0 u , 4xlO u , 5xl0 u , 6xlO u , 7xlO u , 8xl0 u , or 9x10 11 GC per dose including all integers or fractional amounts within the range.
  • the compositions are formulated to contain at least lxlO 12 , 2xl0 12 , 3xl0 12 , 4xl0 12 , 5xl0 12 , 6xl0 12 , 7xl0 12 , 8xl0 12 , or 9xl0 12 GC per dose including all integers or fractional amounts within the range.
  • compositions are formulated to contain at least lxlO 13 , 2xl0 13 , 3xl0 13 , 4xl0 13 , 5xl0 13 , 6xl0 13 , 7xl0 13 , 8xl0 13 , or 9xl0 13 GC per dose including all integers or fractional amounts within the range.
  • the compositions are formulated to contain at least lxlO 14 , 2xl0 14 , 3xl0 14 , 4xl0 14 , 5xl0 14 , 6xl0 14 , 7xl0 14 , 8xl0 14 , or 9xl0 14 GC per dose including all integers or fractional amounts within the range.
  • compositions are formulated to contain at least lxlO 15 , 2xl0 15 , 3xl0 15 , 4xl0 15 , 5xl0 15 , 6xl0 15 , 7xl0 15 , 8xl0 15 , or 9xl0 15 GC per dose including all integers or fractional amounts within the range.
  • the dose can range from lxl0 10 to about lxlO 12 GC per dose including all integers or fractional amounts within the range. All dosages may be measured by any known method, including as measured by oqPCR or digital droplet PCR (ddPCR) as described in, e.g., M. Lock et al, Hum Gene Ther Methods.
  • ddPCR digital droplet PCR
  • an aqueous suspension suitable for administration to an LCA patient comprises an aqueous suspending liquid and about 1 xlO 10 GC or viral particles to about 1 xlO 12 GC or viral particles per eye of a recombinant adeno- associated virus (rAAV) described herein useful as a therapeutic for LCA.
  • rAAV recombinant adeno- associated virus
  • booster dosages of the pharmaceutical compositions of this invention may also be desirable to administer multiple "booster" dosages of the pharmaceutical compositions of this invention. For example, depending upon the duration of the transgene within the ocular target cell, one may deliver booster dosages at 6 month intervals, or yearly following the first administration. The fact that AAV -neutralizing antibodies were not generated by administration of the rAAV vector should allow additional booster administrations.
  • Such booster dosages and the need therefor can be monitored by the attending physicians, using, for example, the retinal and visual function tests and the visual behavior tests described in the examples below. Other similar tests may be used to determine the status of the treated subject over time. Selection of the appropriate tests may be made by the attending physician. Still alternatively, the method of this invention may also involve injection of a larger volume of virus- containing solution in a single or multiple infection to allow levels of visual function close to those found in wildtype retinas.
  • the amount of the vectors, the virus and the replication-defective virus described herein carrying the codon optimized nucleic acid sequences encoding Lebercilin are in the range of about 1.0 x l0 7 VG per eye to about 1.0 x 10 15 VG per eye including all integers or fractional amounts within the range. In one embodiment, the amount thereof is at least lxlO 7 , 2xl0 7 , 3xl0 7 , 4xl0 7 , 5xl0 7 , 6xl0 7 , 7xl0 7 , 8xl0 7 , or 9xl0 7 VG per eye including all integers or fractional amounts within the range.
  • the amount thereof is at least lxlO 8 , 2xl0 8 , 3xl0 8 , 4xl0 8 , 5xl0 8 , 6xl0 8 , 7xl0 8 , 8xl0 8 , or 9xl0 8 VG per eye including all integers or fractional amounts within the range. In one embodiment, the amount thereof is at least lxlO 9 , 2xl0 9 , 3xl0 9 , 4xl0 9 , 5xl0 9 , 6xl0 9 , 7xl0 9 , 8xl0 9 , or 9xl0 9 VG per eye including all integers or fractional amounts within the range.
  • the amount thereof is at least lxlO 10 , 2xl0 10 , 3xl0 10 , 4xl0 10 , 5xl0 10 , 6xl0 10 , 7xl0 10 , 8xl0 10 , or 9xl0 10 VG per eye including all integers or fractional amounts within the range. In one embodiment, the amount thereof is at least lxlO 11 , 2xlO u , 3xl0 u , 4xlO u , 5xl0 u , 6xlO u , 7xlO u , 8xl0 u , or 9xlO u VG per eye including all integers or fractional amounts within the range.
  • the amount thereof is at least lxlO 12 , 2xl0 12 , 3xl0 12 , 4xl0 12 , 5xl0 12 , 6xl0 12 , 7xl0 12 , 8xl0 12 , or 9xl0 12 VG per eye including all integers or fractional amounts within the range. In one embodiment, the amount thereof is at least lxlO 13 , 2xl0 13 , 3xl0 13 , 4xl0 13 , 5xl0 13 , 6xl0 13 , 7xl0 13 , 8xl0 13 , or 9xl0 13 VG per eye including all integers or fractional amounts within the range.
  • the amount thereof is at least lxlO 14 , 2xl0 14 , 3xl0 14 , 4xl0 14 , 5xl0 14 , 6xl0 14 , 7xl0 14 , 8xl0 14 , or 9xl0 14 VG per eye including all integers or fractional amounts within the range. In one embodiment, the amount thereof is at least lxlO 15 , 2xl0 15 , 3xl0 15 , 4xl0 15 , 5xl0 15 , 6xl0 15 , 7xl0 15 , 8xl0 15 , or 9xl0 15 GC per dose including all integers or fractional amounts within the range.
  • the methods comprises dose ranging from lxl0 9 to about lxlO 13 VG per eye per dose including all integers or fractional amounts within the range.
  • the method comprises delivery of the vector in an aqueous suspension.
  • the method comprises administering the rAAV described herein in a dosage of from 1 x 10 9 to 1 x 10 13 GC in a volume about or at least 150 microliters, thereby restoring visual function in said subject. All dosages may be measured by any known method, including as measured by oqPCR or digital droplet PCR (ddPCR) as described in, e.g., M. Lock et al, Hum Gene Ther Methods. 2014 Apr;25(2): 115-25. doi:
  • the volume of carrier, excipient or buffer is at least about 25 ⁇ ⁇ .
  • the volume is about 50
  • the volume is about 75 ⁇ ⁇ .
  • the volume is about 100 ⁇ ⁇ .
  • the volume is about 125 ⁇ ⁇ .
  • the volume is about 150 ⁇ ⁇ .
  • the volume is about 175 ⁇ ⁇ .
  • the volume is about 200 ⁇ In another embodiment, the volume is about 225 ⁇ ⁇ . In yet another embodiment, the volume is about 250 ⁇ ⁇ . In yet another embodiment, the volume is about 275 ⁇ ⁇ . In yet another embodiment, the volume is about 300 ⁇ In yet another embodiment, the volume is about 325 ⁇ In another embodiment, the volume is about 350 ⁇ ⁇ . In another embodiment, the volume is about 375 ⁇ ⁇ . In another embodiment, the volume is about 400 ⁇ ⁇ . In another embodiment, the volume is about 450 ⁇ ⁇ . In another embodiment, the volume is about 500 ⁇ ⁇ . In another embodiment, the volume is about 550 ⁇ ⁇ .
  • the volume is about 600 ⁇ In another embodiment, the volume is about 650 ⁇ ⁇ . In another embodiment, the volume is about 700 ⁇ ⁇ . In another embodiment, the volume is about 800 ⁇ In another embodiment, the volume is between about 150 and 800 ⁇ ⁇ . In another embodiment, the volume is between about 700 and 1000 ⁇ ⁇ . In another embodiment, the volume is between about 250 and 500 ⁇ ⁇ .
  • the viral constructs may be delivered in doses of from at least lxl 0 9 to about least lxlO 11 GCs in volumes of about ⁇ to about 3 ⁇ ⁇ for small animal subjects, such as mice. For larger veterinary subjects having eyes about the same size as human eyes, the larger human dosages and volumes stated above are useful. See, e.g., Diehl et al, J. Applied
  • the lowest effective concentration of virus or other delivery vehicle be utilized in order to reduce the risk of undesirable effects, such as toxicity, retinal dysplasia and detachment.
  • Still other dosages in these ranges may be selected by the attending physician, taking into account the physical state of the subject, preferably human, being treated, the age of the subject, the LCA and the degree to which the disorder, if progressive, has developed.
  • an rAAV carrying the Lebercilin native, modified or codon optimized sequence preferably suspended in a mammalian subject.
  • physiologically compatible carrier may be administered to a desired subject including a human subject.
  • This method comprises administering to a subject in need thereof any of the nucleic acid sequences, expression cassettes, rAAV genomes, plasmids, vectors or rAAV vectors or compositions containing them.
  • the composition is delivered subretinally.
  • the composition is delivered intravitreally.
  • the composition is delivered using a combination of administrative routes suitable for treatment of LCA, and may also involve administration via the palpebral vein or other intravenous or conventional administration routes.
  • the volume and viral titer of each dosage is determined individually, as further described herein, and may be the same or different from other treatments performed in the same, or contralateral, eye.
  • the dosages, administrations and regimens may be determined by the attending physician given the teachings of this specification.
  • the composition is administered in a single dosage selected from those above listed in an affected eye.
  • the composition is administered as a single dosage selected from those above listed in a both affected eyes, either simultaneously or sequentially. Sequential administration may imply a time gap of administration from one eye to another from intervals of minutes, hours, days, weeks or months.
  • the method involves administering the compositions to an eye two or more dosages (e.g., split dosages).
  • a second administration of an rAAV including the selected expression cassette is performed at a later time point. Such time point may be weeks, months or years following the first administration.
  • Such second administration is, in one embodiment, performed with an rAAV having a different capsid than the rAAV from the first administration.
  • the rAAV from the first and second administration have the same capsid.
  • compositions described herein may be delivered in a single composition or multiple compositions.
  • two or more different AAV may be delivered, or multiple viruses [see, e.g., WO 201 1/126808 and WO 2013/049493].
  • multiple viruses may contain different replication-defective viruses (e.g., AAV and adenovirus).
  • non-invasive retinal imaging and functional studies to identify areas of the rod and cone photoreceptors to be targeted for therapy as well as to test the efficacy of treatment.
  • clinical diagnostic tests are employed to determine the precise location(s) for one or more subretinal injection(s).
  • EMG electroretinography
  • perimetry topographical mapping of the layers of the retina and measurement of the thickness of its layers by means of confocal scanning laser ophthalmoscopy (cSLO) and optical coherence tomography (OCT), topographical mapping of cone density via adaptive optics (AO), functional eye exam, Multi-electrode array (MEA), Pupillary Light Responses, etc, depending upon the species of the subject being treated, their physical status and health and the dosage.
  • AO confocal scanning laser ophthalmoscopy
  • OCT optical coherence tomography
  • AO adaptive optics
  • MEA Multi-electrode array
  • Pupillary Light Responses Pupillary Light Responses, etc, depending upon the species of the subject being treated, their physical status and health and the dosage.
  • one or more injections are performed in the same eye in order to target different areas of the affected eye.
  • the volume and viral titer of each injection is determined individually, as further described herein, and may be the same or different from other injections performed in the same, or contralateral, eye. In another embodiment, a single, larger volume injection is made in order to treat the entire eye. In one embodiment, the volume and concentration of the rAAV composition is selected so that only the region of damaged ocular cells is impacted. In another embodiment, the volume and/or concentration of the rAAV composition is a greater amount, in order reach larger portions of the eye, including non-damaged photoreceptors.
  • the method includes performing additional studies, e.g., functional and imaging studies to determine the efficacy of the treatment.
  • additional studies e.g., functional and imaging studies to determine the efficacy of the treatment.
  • such tests include retinal and visual function assessment via electroretinograms (ERGs) looking at rod and cone photoreceptor function, optokinetic nystagmus, pupillometry, water maze testing, light-dark preference, optical coherence tomography (to measure thickness of various layers of the retina), histology (retinal thickness, rows of nuclei in the outer nuclear layer, immunofluorescence to document transgene expression, cone photoreceptor counting, staining of retinal sections with peanut agglutinin - which identifies cone photoreceptor sheaths).
  • ERPs electroretinograms
  • EMGs electroretinograms
  • Other useful post-treatment efficacy test to which the subject is exposed following treatment with a pharmaceutical composition described herein are functional magnetic resonance imaging (fMRI), full-field light sensitivity testing, retinal structure studies including optical coherence tomography, fundus photography, fundus autofluorescence, adaptive optics laser scanning ophthalmoscopy, mobility testing, test of reading speed and accuracy, microperimetry and/or ophthalmoscopy.
  • fMRI functional magnetic resonance imaging
  • fMRI full-field light sensitivity testing
  • retinal structure studies including optical coherence tomography, fundus photography, fundus autofluorescence, adaptive optics laser scanning ophthalmoscopy, mobility testing, test of reading speed and accuracy, microperimetry and/or ophthalmoscopy.
  • a one-time intra-ocular delivery of a composition as described herein is useful in treating LCA in a subject.
  • a one-time intra-ocular delivery of a composition as described herein e.g., an AAV delivery of an optimized LCA5 cassette, is useful in treating LCA in a subject at risk.
  • the composition is administered before disease onset.
  • the composition is administered prior to the initiation of vision impairment or loss.
  • the composition is administered after initiation of vision impairment or loss.
  • the composition is administered when less than 90% of the rod and/or cones or photoreceptors are functioning or remaining, as compared to a non-diseased eye.
  • neonatal treatment is defined as being administered a Lebercilin coding sequence, expression cassette or vector as described herein within 8 hours, the first 12 hours, the first 24 hours, or the first 48 hours of delivery.
  • neonatal delivery is within the period of about 12 hours to about 1 week, 2 weeks, 3 weeks, or about 1 month, or after about 24 hours to about 48 hours.
  • the composition is delivered after onset of symptoms.
  • treatment of the patient e.g., a first injection
  • treatment is initiated prior to the first year of life.
  • treatment is initiated after the first 1 year, or after the first 2 to 3 years of age, after 5 years of age, after 1 1 years of age, or at an older age.
  • treatment is initiated from ages about 4 years of age to about 12 years of age. In one
  • treatment is initiated on or after about 4 years of age. In one embodiment, treatment is initiated on or after about 5 years of age. In one embodiment, treatment is initiated on or after about 6 years of age. In one embodiment, treatment is initiated on or after about 7 years of age. In one embodiment, treatment is initiated on or after about 8 years of age. In one embodiment, treatment is initiated on or after about 9 years of age. In one embodiment, treatment is initiated on or after about 10 years of age. In one embodiment, treatment is initiated on or after about 11 years of age. In one embodiment, treatment is initiated on or after about 12 years of age.
  • treatment can be initiated on or after about 15, about 20, about 25, about 30, about 35, or about 40 years of age.
  • treatment in utero is defined as administering the composition as described herein in the fetus. See, e.g., David et al, Recombinant adeno- associated virus-mediated in utero gene transfer gives therapeutic transgene expression in the sheep, Hum Gene Ther. 201 1 Apr;22(4):419-26. doi: 10.1089/hum.2010.007. Epub 201 1 Feb 2, which is incorporated herein by reference.
  • the composition is readministered at a later date.
  • more than one readministration is permitted.
  • Such readministration may be with the same type of vector, a different viral vector, or via non-viral delivery as described herein.
  • the vector is readministered to the patient to a different portion of the initially injected retina.
  • the vector is readministered to the patient to the same portion of the initially injected retina.
  • any of the above described methods is performed in combination with another, or secondary, therapy.
  • the secondary therapy may be any now known, or as yet unknown, therapy which helps prevent, arrest or ameliorate these mutations or defects or any of the effects associated therewith.
  • the secondary therapy can be administered before, concurrent with, or after administration of the compositions described above.
  • a secondary therapy involves non-specific approaches for maintaining the health of the retinal cells, such as administration of neurotrophic factors, anti-oxidants, anti-apoptotic agents.
  • the non-specific approaches are achieved through injection of proteins, recombinant DNA, recombinant viral vectors, stem cells, fetal tissue, or genetically modified cells. The latter could include genetically modified cells that are encapsulated.
  • a method of generating a recombinant rAAV comprises obtaining a plasmid containing an AAV expression cassette as described above and culturing a packaging cell carrying the plasmid in the presence of sufficient viral sequences to permit packaging of the AAV viral genome into an infectious AAV envelope or capsid.
  • Specific methods of rAAV vector generation are described above and may be employed in generating a rAAV vector that can deliver the codon optimized LCA5 in the expression cassettes and genomes described above and in the examples below.
  • a subject has Leber congenital amaurosis
  • the term “subject” as used herein means a mammalian animal, including a human, a veterinary or farm animal, a domestic animal or pet, and animals normally used for clinical research. In one embodiment, the subject of these methods and compositions is a human. Still other suitable subjects include, without limitation, murine, rat, canine, feline, porcine, bovine, ovine, non-human primate and others. As used herein, the term “subject” is used interchangeably with "patient”.
  • treatment or “treating” is defined encompassing administering to a subject one or more compounds or compositions described herein for the purposes of amelioration of one or more symptoms of LCA.
  • Treatment can thus include one or more of reducing onset or progression of LCA, preventing disease, reducing the severity of the disease symptoms, or retarding their progression, including the progression of blindness, removing the disease symptoms, delaying onset of disease or monitoring progression of disease or efficacy of therapy in a given subject.
  • regulation refers to the ability of a composition to inhibit one or more components of a biological pathway.
  • Example 1 Recombinant rAAV and In Vitro Expression Studies
  • Retinal gene transfer using the most thoroughly studied recombinant AAV serotype, AAV2 has been carried out in >310 eyes in human subjects, and in 29 different clinical trials (clinicaltrials.gov). These trials target diverse diseases including autosomal defects (RPE65 deficiency, retinitis pigmentosa due to MERTK mutations, choroideremia, achromatopsia), a mitochondrial disease (Leber's hereditary optic neuropathy), a complication of age-related macular degeneration (choroidal neovascularization) and end-stage retinal degeneration (using optogenetic therapy).
  • RPE65 deficiency retinitis pigmentosa due to MERTK mutations, choroideremia, achromatopsia
  • a mitochondrial disease Leber's hereditary optic neuropathy
  • choroidal neovascularization choroidal neovascularization
  • end-stage retinal degeneration using optogenetic therapy.
  • Subretinal injection is the
  • AAV2 vectors do not target photoreceptors efficiently, and, as mentioned above, photoreceptors comprise the primary cell type in LCA5 and most other inherited retinal degenerations. For this reason, AAV7m8, a vector generated by evolutionary design was selected. This vector has been shown to target photoreceptors efficiently in diverse species (mice and non-human primates (NHPs)) and using different routes of administration.
  • the efficacy reported herein includes improvements in the ability of treated animals to navigate using visual cues, the restoration of visual pathways to the brain as shown by pupillometry, a reduction in photoreceptor apoptosis, and the preservation of functional photoreceptors with morphology and markers characteristic of this cell type, including presence of rhodopsin in the outer retina.
  • the treated Lca5gt/gt photoreceptors show thick outer nuclear layers with preserved outer segments with stacked outer segment discs. This is in marked contrast to the untreated Lca5gt/gt retinas, which were reduced to a single row of non-contiguous photoreceptor nuclei by 3 months of age 19. The improvements were not permanent.
  • LCA5 gene therapy is capable of at least partially restoring responses mediated by both rod and cone photoreceptors.
  • the responses of the cells have near normal kinetics, including responses reflecting the activity of a variety of ganglion cell types as well as a reversal of the dominating melanopsin responses observed in the untreated Lca5gt/gt retinas.
  • the results are complementary with the pupillometry and visual behavior findings and will provide the framework for future studies aiming to further characterize and optimize the treatment effects (including studies of visual behavior in low illuminance conditions).
  • Recombinant AAVs were generated in the Center for Advanced Retinal and Ocular
  • the human (h) wildtype lebercilin-encoding optimized cDNA (hopt.ZC45) was custom-designed for optimal codon usage and synthesized by DNA2.0 (Menlo Park, CA).
  • the LCA5 cDNA was placed under the control of a hybrid chicken ⁇ -actin (CBA) exon 1 flanked by intron 1 sequences and with cytomegalovirus (CMV) immediate early enhancer (FIG. 1A and IE).
  • CBA chicken ⁇ -actin
  • CMV cytomegalovirus
  • FOG. 1A and IE immediate early enhancer
  • a bovine growth hormone poly(A) followed the cDNA.
  • a long staffer sequence was included to prevent reverse packaging (i.e. of non-transgene containing) vector from the AAV2 inverted terminal repeats.
  • the vectors were made by triple transfection and formulated in excipient consisting of phosphate-buffered saline (PBS) containing and 0.001% Pluronic F68 (PF68). See, e.g. Mizukami, Hiroaki, et al. A Protocol for AAV vector production and purification. Diss. Di-vision of Genetic Therapeutics, Center for MolecularMedicine, 1998. Control vectors incorporated the enhanced green fluorescent protein (eGFP) cDNA in place of the LCA5 cDNA.
  • PBS phosphate-buffered saline
  • PF68 Pluronic F68
  • the rAAVs were tested for expression of the appropriate sized transgenic protein by Western blot.
  • 8431 cells were plated at 2 x 10 6 cells per well. Two days after plating, cells were transduced with AAV7m8.hopt.LCA5 (or AAV7m8.CBA.EGFP as control) at 1 x 10 5 or 5 x 10 5 vg. 48 hours later, cells were harvested and processed for electrophoresis and Western blot.
  • Antibodies include an LCA5 rabbit polyclonal antibody (Proteintech, Rosemont, IL) and the signals are quantified, with each value is corrected for background and protein loading differences through normalization with the GAPDH immunosignal.
  • the AAV7m8.CBA.hopt.LCA5 virus is able to drive efficient expression of the LCA5 transgene in 8431 cells.
  • Production of the predicted ⁇ 8 lkDA Lebercilin protein after infection with AAV7m8.CBA.hopt.LCA5 demonstrates a dose-dependent responses.
  • Example 2 LCA mouse model Lca5-/- Mouse Studies Development of a proof-of-concept of gene augmentation therapy in the Lca5gt/gt mouse model entails several challenges: 1) because retinal degenerative changes begin very early and progress rapidly, intervention must be carried out in neonatal mice; 2) since this is a
  • AAV7m8 designed by directed evolution, to deliver a codon optimized human lebercilin-encoding cDNA.
  • Lebercilin localizes to connecting cilia of photoreceptor cells (22).
  • the connecting cilium is a transition zone between the photoreceptor cell body inner segment and the antennae-like outer segments that supports selective transport of proteins and membrane vesicles.
  • the connecting cilium is thus the conduit supporting bi-directional protein trafficking along ciliary microtubule tracks, or intraflagellar transport (IFT).
  • LCA5 mutations interfere with IFT,(22) thereby causing an early-onset defect in photoreceptor outer segment development and a failure to correctly traffic two different proteins expressed specifically in photoreceptors, arrestin and opsin.
  • Knockout of the Lca5 gene in mice resulted in a retinal degeneration phenotype.
  • the Lca5- /- mice develop patches of de-pigmented retina, never develop outer segments and lack cone and rod ERG responses to light. There is an early and rapidly progressive retinal degeneration and only a single (sickly) row of nuclei is present in the ONL by 2 months of age. (22)
  • the Lca5-/- mice were utilized as an animal model for LCA.
  • mice were purchased from Jackson Labs (Bar Harbor, ME) and a line was generated by brother-sister crossings. Verification of the genotype of all animals used in the study was performed (see Supplementary Methods). Mice were on a 12-hour light/12-hour dark cycle, and food/water was provided ad libitum. The studies were performed in compliance with federal and institutional regulations. Subretinal injections were carried out unilaterally in neonatal mice as described previously (28) in cohorts of pups. Anesthesia in mice at postnatal day 5 (PN5) was hypothermia. At postnatal day 15 (PN15), animals were anesthetized with ketamine/xylazine. Table 1 shows the number of animals used per cohort.
  • AAV7m8.CBA.hopt.LCA5 was injected at a total of 9.20 x 10 9 vg in 1 ⁇ in cohort 1 (Table 1).
  • the injection solution contained 5% v/v of AAV7m8.CBA.EGFP so that the area of injection could be identified with certainty at later time points through presence of enhanced green fluorescent protein (EGFP).
  • Additional animals received sham injection (cohort 2), received injection of AAV7m8.CBA.EGFP alone (cohort 3), or were maintained uninjected as controls (cohort 4). After injection, pups were returned to their mothers until the time of weaning.
  • Table 1 Cohorts of neonatal Lca5-/- mice injected at the designated postnatal day (PN) and studied in vivo. ROA - route of administration.
  • Ophthalmoscopy was carried out about 1 month post injection to verify that media were clear, retinas were not detached, and thus that there had been no surgical complications. Any animals that had corneal or vitreal opacities were excluded from further study.
  • mice Cohorts of mice were bred and studied, and injections were carried out at early postnatal
  • PN5 juvenile (PN 15) time points (Table 1). Injections were found largely without complications. After injections at PN5, the majority of the animals were found to be free of corneal or vitreal opacities that would interfere with further testing. The few animals with opacities were excluded from further analyses.
  • the AAV7m8.C A.hopt-LCA5 virus was able to drive efficient expression of the human
  • Immunofluorescence analysis shows presence of lebercilin protein in the ONL, inner segments (IS) and connecting cilia (CC) in wild type adult retina ( Figure IE).
  • Tests of visual and retinal function included a light-cued water maze (2-3 months post injection), pupillometry (2.5 months post injection), multi-electrode array (3 months post injection) and electroretinography utilizing the mice indicated or described in Example 2.
  • Retinas were then evaluated for histopathology and immunofluorescence as described in Example 4. A thorough tissue analysis was also followed.
  • A. Electroretinography We recorded ERGs from 8 Lca5gt/gt mice, 5 wild-type animals, and 16 Lca5gt/gt mice whose left eyes had been injected with excipient and the right eyes with AAV7m8.
  • C A.hopt- LCA5 on PN5 intravitreally. None of the eyes of Lca5gt/gt uninjected controls or Lca5gt/gt eyes injected with excipient (a total of 32 eyes) produced detectable ERG responses. Out of 16 eyes injected with AAV7m8.
  • CpA.hopt-LCA5 four showed ERG responses to dim flashes of light in the dark-adapted state consistent with rod-mediation of the signals, as well as mixed rod-cone responses to brighter flashes of light that resembled a smaller, scaled version of wild-type (WT) mixed rod-cone ERG waveforms ( Figure S2).
  • WT wild-type
  • Figure S2 Light-adapted ERGs were also detectable in these eyes suggesting cone-mediation of the photopic responses.
  • Detectable ERGs post-treatment ranged in amplitude from 20 to 25% the WT amplitudes ( Figure S2). The results suggest restoration of both rod and cone photoreceptor function after gene therapy in some of the animals.
  • Water maze testing was performed to evaluate each animal's ability to identify the chamber containing a submerged platform in a 5-chamber water maze (FIG. 11).
  • the apparatus was maintained in a room without extraneous light and the light source was placed directly over the platform prior to the test.
  • the dark-adapted mouse was released in the center of the maze and given 60 seconds, without interruption, to find and place all four paws on the lit platform.
  • Training was carried out in room light (about 200 Lux) prior to dark adapting the mice and carrying out the test under dim light procedures. During the training, if the mouse was unable to find the platform at the end of the 60 seconds, the experimenter guided the mouse to the platform. For each trial, the light and platform were placed in a different chamber using random selection.
  • Training pass criteria were defined as the ability of the mouse to independently enter the correct chamber, without deviating into a different chamber, and mount the platform within 60 seconds in more than 5 of 9 sequential trials. All mice were trained for 5 days regardless of the day that training pass criteria were met. Testing was carried out for 4 days using the same procedure as that used in training but with a series of filters to further dim the light source.
  • the luminances were: 1.06 xlO 5 , 8.69 x 10 3 , 5.87 x 10 2 scot cd m 2 , or with the light off, the luminance was 0 scot cd m 2 .
  • Animals were trained and then tested with the light-cued water maze at 2-2.5 months of age. Light-cued water maze test results showed that the AAV7m8.CBA.hopt.LCA5 subretinally or intravitreally injected groups performed significantly better than controls. (Table 2B; see values in BOLD; FIG. 3).
  • Table 2A shows the raw data, which are numbers of animals analyzed in each cohort and then tested under the designated light levels using the water maze.
  • Table 2B shows the results of statistical analyses using one-way analysis of variance (ANOVA). The treated animals were able to pass the test at 8.69 x 10 3 scot cd m 2 whereas animals from the control excipient-injected cohort were not (p ⁇ 0.01). Table 2. Results of water maze testing in Lca5-/- mice.
  • AAV7m8-hopt-LCA5 0.24 0.01 0.46 0.74 intravitreal at PN5 vs.
  • AAV7m8-hopt-LCA5 0.37 0.46 0.69 0.48 intravitreal at PN15 vs.
  • the amplitudes of pupillary constriction were measured during a series of 10 light flashes per eye. Flash intensity was 1,000 scot lux. PLRs were defined as having a maximum amplitude of pupil constriction response within the 0.6- 1.2 second interval following the flash that was more than 3 standard deviations of the pre-stimulus diameter.
  • a scientist masked to the treatment paradigm evaluated the data from each animal prior to analyses to be sure that each pupil was tracked accurately. If not, that particular animal was excluded from further analyses.
  • contralateral sham-injected control left Lca5-/- eyes were compared at each light intensity. By dividing the normalized amplitude of constriction of the treated eye with that of the untreated eye, the percentage of the Maximum Differential Amplitude of Response (MDAR) could be obtained. Higher percentage of the MDAR indicated a stronger response to intervention. Additional age- matched untreated Lca5-/- and wildtype (C57B1/6) mice were used as positive and negative controls, respectively. The MDARs of untreated Lca5-/- and C57B1/6 mice were about 0 since there was minimal difference between the two eyes.
  • MDAR Maximum Differential Amplitude of Response
  • AAV7m8.hopt-LCA5 compared to untreated control Lca5-/- animals (FIG. 2).
  • PLR Pupillary Light Reflex
  • Multi-electrode array (MEA) testing was carried out on 5 animals 2.5-3.5 months after they had received unilateral intravitreal injection with AAV7m8.CMV/CBA.hopt.LCA5 (about 9.87 x 10 10 vg/eye) spiked with 5% (v/v) AAV7m8.CBA.GFP. Contralateral eyes were sham- injected and used as negative controls. Five untreated age-matched wildtype (WT) C57B1/6 mice served as positive controls. Retinas from light-adapted mice were dissected under red light and mounted ganglion cell side down in the perforated MEA chamber. Presence of GFP in the explants confirmed exposure of photoreceptors to AAV.
  • WT age-matched wildtype
  • MEA measures the output signal of the retina sent to the brain and thus, in addition to testing photoreceptor function, also provides information about retinal wiring.
  • Lca5gt/gt retinas Responses at the brighter end of intensities should originate in cones (the brightest intensity should be more than capable of driving both M- and S-cones by producing around 1.00e9 and 6.00e6 photoisomerizations per second per cell in M- and S-cones respectively).
  • the brightest intensity should be more than capable of driving both M- and S-cones by producing around 1.00e9 and 6.00e6 photoisomerizations per second per cell in M- and S-cones respectively.
  • the brightest stimulation series after exposure to the brightest stimulation series at the end of the 1st intensity series run (light sensitive retinas were subjected to at least 2 intensity series runs ranging from the dim scotopic to the brightest photopic intensities in -0.5 log increments), scotopic responses disappeared, but photopic responses were not significantly affected (Figure 5 C and D).
  • WT /light responsive 5 intensity range from 40 to 2e9 photons per um A 2 per second
  • Retinas from sham-injected contralateral eyes showed minimal to abolished responses (Data not shown. Control Lca5-/-).
  • Responses in AAV7m8.hopt.LCA5-treated retinas were similar to those in retinas of wildtype mice. Flicker responses showing that after exposure to the brightest light
  • PLR Pupillary light reflexes
  • AAV7m8.C A.hopt-LCA5 injected Lca5gt/gt animals performed better than uninjected controls in a statistically significant manner under at least one light condition.
  • Table 2; Figure 3 The animals treated subretinally at PN5 showed a significantly improved pass rate at every lighting test condition (1.06E+05, 8.69E+03, and 5.87E+02 scot cd m-2), than control excipient-injected cohort (p ⁇ 0.01).
  • IVT or SR improvements were minimal (Figure 3).
  • mice used only the given light cue testing was performed under no light condition (0.00 scot cd m2) and over 90% of mice failed the test (including normal-sighted mice; Figure 3B).
  • Example 4 Ocular Histology, Histopathology, Immunofluorescence and TU EL Assay
  • TEM Transmission electron microscopy
  • Micrographs of hematoxylin and eosin-stained (H&E) retina showed that inner/outer segments (IS/OS) and ONL are preserved through the 3 months timepoint (PN99) after either IVT or SR injection of AAV7m8.hopt-LCA5 (Data not shown).
  • sham-injected control retinas lack such layers at PN99 (Data not shown).
  • the thickness of photoreceptor cell layers in retinas that were treated by IVT or SR injection with AAV7m8.C A.hopt-LCA5 at PN5 was significantly greater than in control retinas (FIG. 4). There was also a noticeable border in thickness between the areas of retinas exposed to or unexposed to AAV in retinas treated by subretinal injection (Data not shown). The preservation of photoreceptor layers persisted through the latest timepoint (PN99). The increased thickness was due to an increased number of rows in the outer nuclear layer, and presence of inner and outer segments (FIG. 4). Consistent with this, there was a much
  • TEM Transmission electron microscopy
  • AAV7m8.hopt.LCA5-treated retinas showed elaboration of outer segments complete with stacked outer segment disks, 9+0 microtubule arrays typical of primary cilia (FIG. 7B) and connecting cilia (FIG 7).
  • untreated Lca5-/- retinas lacked both connecting cilia and outer segments.
  • the untreated retinas lack outer segments and show massive degeneration of photoreceptors with only pyknotic nuclei and remnants of photoreceptor organelles. Only disorganized, degenerating photoreceptor cell bodies were present in control untreated or sham-injected retinas.
  • AAV7m8.hopt-LCA5 resulted into normal translocation patterns of phototransduction proteins into outer segments. Arrestin translocates properly after light exposure in AAV7m8.hopt-LCA5- treated retinas. Such activity cannot be assessed in the control retinas due to the degeneration of IS/OS.
  • the data reflect restoration of the intraflagellar transport defect in the mouse retina after delivery of the wild type (WT) hLCA5 cDNA.
  • AAV7m8 was used to test for efficacy after delivery of the native human Lebercilin-encoding cDNA.
  • This vector has been shown by others to efficiently target photoreceptors after intra-vitreal delivery. (25-27).
  • AAV7m8 By using AAV7m8 to deliver the hLCA5 cDNA to the diseased photoreceptors early in life, gene augmentation therapy resulted in both structural and functional improvement of the Lca5-/- mouse retina and of vision.
  • the efficacy reported in the present example included the improved ability of this model to navigate using visual cues, restoration of rod and cone photoreceptor responses as shown by Multi-electrode array (MEA), a reduction in apoptotic cell death (and thus preservation of photoreceptors), and presence of cell biologic and physical features characteristic of normally functioning photoreceptors, such as presence of rhodopsin in the outer retina and development of normal-appearing outer segments, with stacked outer segment discs.
  • MEA results showed that given successful injection and enough post injection time to express the transgenic protein and restore function of rod/cone outer segments, gene therapy restored degenerated retinal cells to a state nearly indistinguishable from the WT conditions.
  • P3000 pupillometer and PupilFit6 software (Monmouthshire, UK). Pupillary responses to light were recorded after presentation of 10 lux green light stimuli to the right eye for 0.2 seconds followed by dark intervals.
  • An infrared sensitive camera that captures video images at 25 frames per second, allowing the pupil diameter of both eyes to be measured every 40 ms.
  • Pupillary light reflex testing was carried out to determine whether there was any evidence of function in the residual photoreceptors present in an adult with homozygous LCA5 mutations. As shown in FIG 8, pupillary light reflexes were present in this individual with the same temporal sequence as those in a normal-sighted individual. However, the amplitudes of response were diminished considerably compared to the normal-sighted individual.
  • iPSC human induced pluripotent stem cell
  • PBMCs Peripheral blood monocytes
  • JB605 was a compound heterozygote for LCA5 Gln279Stop CAG>TAG het and
  • the other, JB590 was homozygous for LCA5 C.8350T p.Q279X.
  • the wildtype cells were from individual PBWT4.6.
  • Induced pluripotent stem cell lines were generated from each of the three individuals.
  • PBMCs were cultured in expansion media consisting of QBSF-60 (Invitrogen, Carlsbad, CA) media supplemented with cytokines and hormone. The media was replenished every 2-3 days for a period of 7-9 days until the cells entered a stage of exponential growth.
  • expanded PBMCs were transduced with the rTTA lentivirus and doxycycline inducible "stem cell cassette" in the lentivirus vector delivering OCT4, KLF4, SOX2, and cMyc cDNA and microRNA 302/367 cluster driven by the TetO/CMV promoter.
  • Cells were grown in expansion media supplemented with polybrene (5 ⁇ g/ ml; Sigma-Aldrich, St. Louis, MO). The cells were incubated for 20-24 h at 37°C. Infected cells were then washed, and placed in expansion media supplemented with 1 ⁇ g/ml of doxycycline (DOX).
  • polybrene 5 ⁇ g/ ml
  • DOX doxycycline
  • IMDM Iscove's modified Dolbecco's medium
  • FBS fetal bovine serum
  • penicillin streptomycin penicillin streptomycin
  • L-glutamine penicillin streptomycin
  • beta-mercaptoethanol nonessential amino acids
  • DOX basic fibroblast growth factor
  • MEF plates mouse embryonic fibroblast plates
  • hESC human embryonic stem cell
  • DMEM/F 12 20% knockout serum replacement, nonessential amino acid, 4 ng/ml bFGF, 0.001% beta-mercaptoethanol, penicillin/streptomycin, L-glutamine, and 1 ⁇ g/ml of DOX (Invitrogen, Carlsbad, CA).
  • iPSC-like colonies were manually picked and expanded on matrigelcoated MEF plates for 6 passages, then transitioned to 0.1% gelatin-coated MEF plates for a minimum of 15 passages.
  • iPSCs Characterization of iPSCs was based on surface antigen expression measured by flow cytometry using antibodies against SSEA3+, SSEA4+, TRA-1-60, and TRA- 1-81 (Biolegend, San Diego, CA), and RT-qPCR analysis included pluripotency expression markers: DMNT3B, ABCG2, REX1, OCT4, SOX2, NANOG, cMYC, KLF4.
  • Karyotyping of iPSCs was carried out by G banding to produce a visible karyotype. The ability of the cells to differentiate into multiple different lineages was carried out using a PCR germ layer assay (Qiagen. Germantown, MD, USA).
  • the characterized iPSCs were differentiated into RPE by activation of the Wnt signaling pathway, inhibition of the fibroblast growth factor signaling pathway, and inhibition of the Rho- associated, coiled-coil containing protein kinase signaling pathway.
  • Expanded pigmented cells were purified by plate adhesion on 8-chamber slides on geltrex matrix. Enriched cells were cultured until they developed a cobblestone appearance with cuboidal shape.
  • the characteristics of iPS-RPE were confirmed by gene expression, immunocytochemistry, and microscopy.
  • Cilia lengths were measured after fixing and staining for ARL 13B and Pericentrin. Cilia lengths were measured in three dimensions using custom-designed software designed by the Wistar Imaging Facility (Wistar Institute, Philadelphia, PA)
  • Cilia are evaluated in the control and LCA5 iPSC-RPE cell lines for density and length. There are significantly fewer cilia present on the RPE cells derived from the two LCA5 patients than on the wildtype cell line. In addition, the cilia on the LCA5-derived lines are significantly shorter than those in the wildtype cells.
  • LCA5 patients can retain photoreceptors including in the foveal outer nuclear layer, for up to 3 decades. (21, 29). Since successful gene therapy requires that the cells be present (even if they are sickly), LCA5 photoreceptors may be amenable to treatment. The fact that the instant examples demonstrated that retained photoreceptors in an adult with LCA5 showed a similar temporal pattern of light responsiveness (albeit reduced amplitude) as photoreceptors from a normal-sighted individual is encouraging. These results further supported the fact that the residual photoreceptors in these abnormal retinas were viable despite their structural and functional deficits. Further, the cilia from the LCA patient-derived iPSC-RPE cells were significantly shorter than those from the wildtype control cells.
  • Genomic DNA was PCR-amplified using the following oligos: LCA- 13F6 (common F) GCCTGTTCCTGCTTGCTTAC (enclosed as SEQ ID NO: 13); LCA- 13R2 (WT Reverse) TGCTTTCCAAAGTAAGCACAAA (enclosed as SEQ ID NO: 14)
  • PCR was run for 35 cycles with an annealing temperature of 50°C and an extension step at 72°C.
  • the predicted products are 353 bp (wildtype) and 439 bp (Lca5-/-).
  • Example 8 Intravitreal injection of AAV7m8.hLCA5 restores photoreceptor function in Lca5-/- mice to nearly WT levels
  • LCA5 Early onset vision loss results from mutations in LCA5, a gene which encodes Lebercilin, a protein critical for the health and function of photoreceptors. Because there can be relative preservation of photoreceptors, LCA5 disease may be amenable to gene augmentation therapy. The possibility that gene augmentation therapy using intravitreal delivery of
  • AAV7m8.CMV/CBA.hLCA5 restores photoreceptor and retinal function using multi electrode array (MEA) in an Lca5 mouse model (Lca5-/-) was tested.
  • MEA multi electrode array
  • AAV7m8.CMV/CBA.hLCA5 (approximately 9.87 x 10 10 vg/eye) mixed with 5% (v/v) AAV7m8.CBA.GFP.
  • Contralateral eyes were uninjected and used as negative control. 3 months after the intervention and under light adapted conditions, Lca5-/- retinas were dissected under red light and mounted ganglion cell side down in the perforated MEA chamber. The same dissection and preparation procedure was done for untreated age-matched wild type mice (C57B1/6) as a positive control. Presence of GFP confirmed exposure of photoreceptors to the AAV.
  • All ganglion cell types identifiable in the WT retinas under full field stimulation were detected in the Lca5-/- treated retinas after spike sorting.
  • treated Lca5-/- retinas were responsive to 4Hz flicker stimulation in the intensity range of 3.53 x 10 2 -2.00 x 10 9 photons x s "1 x ⁇ "2 .
  • Only one of the 7 control untreated Lca5-/- retinas demonstrated very weak light responses at the bright photopic stimuli likely indicative of the remaining cone function.
  • All untreated Lca5-/- retinas demonstrated slow melanopsin driven responses at brighter intensities which were absent in the light sensitive treated Lca5-/- retinas.
  • gene therapy restores degenerated retinal cells to a state nearly indistinguishable from the WT conditions.
  • LCA itself is one of the most severe retinal degenerative diseases
  • LCA5 is one of the most severe subtypes within this category.
  • LCA5 patients enjoy only light perception early in life, and it is difficult even to carry out structural studies and specialized tests of visual function in these ultra-low vision subjects due to nystagmus. Thus, this disease is considered difficult to approach.
  • the phenotype of the Lca5 -/- mouse reflects many of the clinical findings in humans with LCA5 mutations. The rescue data in the Lca5-/- mouse provide hope that a similar gene augmentation approach in humans could result in improved vision.
  • PLR testing was carried out to determine whether there was any evidence of function in the residual photoreceptors present in human adult with homozygous LCA5 mutations. As shown in Figure S I 1, PLRs were present in this individual with the same temporal sequence as those in a normal-sighted individual. However, the amplitudes of response were diminished considerably compared to the normal-sighted individual.
  • Example 10 Loss of Lebercilin causes dysregulation of RPE maturation and ciliary function in cellular and animal models
  • LCA5 retinal pigmented epithelium
  • Immunostaining also identified differences in the epithelial barrier consistent with altered maturation due to loss of LCA5 protein function.
  • Jacobson SG Cideciyan AV, Ratnakaram R, Heon E, Schwartz SB, Roman AJ, et al.
  • Cideciyan AV Jacobson SG
  • Beltran WA Sumaroka A
  • Swider M Iwabe S, et al.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)

Abstract

L'invention concerne des compositions et méthodes pour le traitement de l'amaurose congénitale de Leber (ACL) chez un sujet. Selon un aspect, l'invention concerne un vecteur viral adéno-associé qui comprend une molécule d'acide nucléique comprenant une séquence codant pour la Leberciline. Dans un autre aspect, la léberciline a une séquence d'acides aminés de SEQ ID NO: 1. Dans encore un autre aspect, la molécule d'acide nucléique a une séquence de SEQ ID NO: 3 ou un variant de celle-ci. Dans les modes de réalisation souhaités, le sujet est un homme, un chat, un chien, un mouton, ou un primate non humain.
PCT/US2018/020470 2017-03-01 2018-03-01 Thérapie génique pour troubles oculaires WO2018160849A1 (fr)

Priority Applications (14)

Application Number Priority Date Filing Date Title
RU2019130004A RU2019130004A (ru) 2017-03-01 2018-03-01 Генная терапия при глазных заболеваниях
US16/489,770 US11564996B2 (en) 2017-03-01 2018-03-01 Gene therapy for ocular disorders
AU2018228881A AU2018228881B2 (en) 2017-03-01 2018-03-01 Gene therapy for ocular disorders
BR112019017327A BR112019017327A2 (pt) 2017-03-01 2018-03-01 terapia gênica para distúrbios oculares
JP2019547457A JP7211960B2 (ja) 2017-03-01 2018-03-01 眼疾患の遺伝子治療
CN201880029279.2A CN110582572A (zh) 2017-03-01 2018-03-01 用于眼部病症的基因疗法
CA3054136A CA3054136A1 (fr) 2017-03-01 2018-03-01 Therapie genique pour troubles oculaires
EP18760861.7A EP3589738A4 (fr) 2017-03-01 2018-03-01 Thérapie génique pour troubles oculaires
KR1020197027823A KR102545070B1 (ko) 2017-03-01 2018-03-01 안구 장애에 대한 유전자 치료
KR1020237020037A KR20230093072A (ko) 2017-03-01 2018-03-01 안구 장애에 대한 유전자 치료
IL26894619A IL268946A (en) 2017-03-01 2019-08-27 Gene therapy for ocular disorders
US18/061,633 US20230233709A1 (en) 2017-03-01 2022-12-05 Gene therapy for ocular disorders
JP2023002799A JP2023040219A (ja) 2017-03-01 2023-01-12 眼疾患の遺伝子治療
AU2024202693A AU2024202693A1 (en) 2017-03-01 2024-04-24 Gene Therapy For Ocular Disorders

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762465649P 2017-03-01 2017-03-01
US62/465,649 2017-03-01
US201762469642P 2017-03-10 2017-03-10
US62/469,642 2017-03-10

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/489,770 A-371-Of-International US11564996B2 (en) 2017-03-01 2018-03-01 Gene therapy for ocular disorders
US18/061,633 Continuation US20230233709A1 (en) 2017-03-01 2022-12-05 Gene therapy for ocular disorders

Publications (2)

Publication Number Publication Date
WO2018160849A1 true WO2018160849A1 (fr) 2018-09-07
WO2018160849A8 WO2018160849A8 (fr) 2019-10-03

Family

ID=63370488

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/020470 WO2018160849A1 (fr) 2017-03-01 2018-03-01 Thérapie génique pour troubles oculaires

Country Status (11)

Country Link
US (2) US11564996B2 (fr)
EP (1) EP3589738A4 (fr)
JP (2) JP7211960B2 (fr)
KR (2) KR20230093072A (fr)
CN (1) CN110582572A (fr)
AU (2) AU2018228881B2 (fr)
BR (1) BR112019017327A2 (fr)
CA (1) CA3054136A1 (fr)
IL (1) IL268946A (fr)
RU (1) RU2019130004A (fr)
WO (1) WO2018160849A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022165313A1 (fr) 2021-02-01 2022-08-04 Regenxbio Inc. Thérapie génique de céroïdes-lipofuscinoses neuronales
US20220387627A1 (en) * 2021-06-03 2022-12-08 Hope for Hasti Vectors and gene therapy for treating cornelia de lange syndrome

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20240014102A (ko) * 2018-06-29 2024-01-31 우한 뉴로프스 바이오테크놀로지 리미티드 컴퍼니 레버 유전성 시신경병증의 치료를 위한 조성물 및 방법

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160032319A1 (en) * 2013-03-15 2016-02-04 The Children's Hospital Of Philadelphia Vectors comprising stuffer/filler polynucleotide sequences and methods of use
US20160263246A1 (en) * 2001-04-13 2016-09-15 The Trustees Of The University Of Pennsylvania Method of Treating or Retarding the Development of Blindness

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5436146A (en) 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
US6268213B1 (en) 1992-06-03 2001-07-31 Richard Jude Samulski Adeno-associated virus vector and cis-acting regulatory and promoter elements capable of expressing at least one gene and method of using same for gene therapy
US5869305A (en) 1992-12-04 1999-02-09 The University Of Pittsburgh Recombinant viral vector system
US6204059B1 (en) 1994-06-30 2001-03-20 University Of Pittsburgh AAV capsid vehicles for molecular transfer
US5741683A (en) 1995-06-07 1998-04-21 The Research Foundation Of State University Of New York In vitro packaging of adeno-associated virus DNA
US6093570A (en) 1995-06-07 2000-07-25 The University Of North Carolina At Chapel Hill Helper virus-free AAV production
ES2224375T3 (es) 1997-04-14 2005-03-01 Cell Genesys, Inc. Metodos para aumentar la eficacia del producto de aav recombinante.
EP1080218A1 (fr) 1998-05-27 2001-03-07 University of Florida Procede de preparation de compositions de virus adeno-associes de recombinaison a l'aide d'un gradient d'iodixananol
JP2002538770A (ja) 1998-11-10 2002-11-19 ユニバーシティ オブ ノース カロライナ アット チャペル ヒル ウイルスベクターとその製造及び投与の方法
ATE403715T1 (de) 1999-08-09 2008-08-15 Targeted Genetics Corp Erhöhung der expression einer einzelsträngigen, heterologen nukleotidsequenz von einem rekombinanten viralen vektor durch ausgestaltung der sequenz in einer art und weise, dass basenpaarungen innerhalb der sequenz entstehen
AU6814901A (en) 2000-06-01 2001-12-11 Univ North Carolina Methods and compounds for controlled release of recombinant parvovirus vectors
DK1310571T3 (da) 2001-11-13 2006-06-19 Univ Pennsylvania Fremgangsmåde til identifikation af ukendte adeno-associerede virussekvenser (AAV-sekvenser) og et kit til fremgangsmåden
EP1453547B1 (fr) 2001-12-17 2016-09-21 The Trustees Of The University Of Pennsylvania Sequences du serotype 8 du virus associe a l'adenovirus (aav), vecteurs les contenant et utilisations correspondantes
WO2003104413A2 (fr) 2002-06-05 2003-12-18 University Of Florida Production de virions de virus adeno-associe (aav) recombinants pseudo-types
EP1486567A1 (fr) 2003-06-11 2004-12-15 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Vecteur derivé d'un virus associé aux adenovirus pour la thérapie génique
US8005620B2 (en) 2003-08-01 2011-08-23 Dna Twopointo Inc. Systems and methods for biopolymer engineering
EP2434420A3 (fr) 2003-08-01 2012-07-25 Dna Twopointo Inc. Systèmes et procédés d'ingénierie de biopolymère
ES2411479T3 (es) 2003-09-30 2013-07-05 The Trustees Of The University Of Pennsylvania Clados de virus adeno-asociados (AAV), secuencias, vectores que los contienen, y usos de los mismos
EP3085389A1 (fr) 2005-04-07 2016-10-26 The Trustees Of The University Of Pennsylvania Procédé d'augmentation de la fonction d'un vecteur aav
EP1777906A1 (fr) 2005-06-09 2007-04-25 Matsushita Electric Industrial Co., Ltd. Appareil de compensation d erreur d amplitude et appareil de compensation d erreur d orthogonalité
EP2007795B1 (fr) 2006-03-30 2016-11-16 The Board Of Trustees Of The Leland Stanford Junior University Protéines de capsides aav
EP2091966A2 (fr) * 2006-12-01 2009-08-26 Oxford BioMedica (UK) Limited Construction promotrice
US8969353B2 (en) 2008-11-07 2015-03-03 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof
US9315825B2 (en) 2010-03-29 2016-04-19 The Trustees Of The University Of Pennsylvania Pharmacologically induced transgene ablation system
JP5922095B2 (ja) 2010-03-29 2016-05-24 ザ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア 薬理学的に誘導される導入遺伝子アブレーション系
US9249425B2 (en) * 2011-05-16 2016-02-02 The Trustees Of The University Of Pennslyvania Proviral plasmids and production of recombinant adeno-associated virus
IL280771B2 (en) 2011-06-08 2024-03-01 Shire Human Genetic Therapies Preparations of lipid nanoparticles and methods for administration of mRNA
EP3800254A1 (fr) 2012-06-08 2021-04-07 Ethris GmbH Administration pulmonaire d'arn messager
WO2014011210A1 (fr) 2012-07-11 2014-01-16 The Trustees Of The University Of Pennsylvania Thérapie génique induite par vaa s'appliquant à la dégénérescence rétinienne liée à l'x de rpgr
US9567376B2 (en) 2013-02-08 2017-02-14 The Trustees Of The University Of Pennsylvania Enhanced AAV-mediated gene transfer for retinal therapies
ES2697249T3 (es) * 2013-03-11 2019-01-22 Fond Telethon miR-204 y miR-211 y usos de los mismos
US9158889B2 (en) * 2013-04-26 2015-10-13 Oral4D Systems Ltd. Electronic dental charting
WO2015012924A2 (fr) 2013-04-29 2015-01-29 The Trustees Of The University Of Pennsylvania Cassettes d'expression préférentielle pour tissus modifiées par un codon, vecteurs les contenant, et utilisation
US10392622B2 (en) * 2014-08-01 2019-08-27 The Trustees Of The University Of Pennsylvania Compositions and methods for self-regulated inducible gene expression

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160263246A1 (en) * 2001-04-13 2016-09-15 The Trustees Of The University Of Pennsylvania Method of Treating or Retarding the Development of Blindness
US20160032319A1 (en) * 2013-03-15 2016-02-04 The Children's Hospital Of Philadelphia Vectors comprising stuffer/filler polynucleotide sequences and methods of use

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEN, X ET AL.: "Next-generation Sequencing Extends the Phenotypic Spectrum for LCA5 Mutations: Novel LCA5 Mutations in Cone Dystrophy", SCIENTIFIC REPORTS, vol. 6, no. 24357, 12 April 2016 (2016-04-12), pages 1 - 6, XP055538213 *
GEORGIADIS, A ET AL.: "Development of an optimized AAV2/5 gene therapy vector for Leber congenital amaurosis owing to defects in RPE65", GENE THERAPY, vol. 23, no. 12, December 2016 (2016-12-01), pages 857 - 862, XP055538211 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022165313A1 (fr) 2021-02-01 2022-08-04 Regenxbio Inc. Thérapie génique de céroïdes-lipofuscinoses neuronales
US20220387627A1 (en) * 2021-06-03 2022-12-08 Hope for Hasti Vectors and gene therapy for treating cornelia de lange syndrome

Also Published As

Publication number Publication date
AU2024202693A1 (en) 2024-05-16
US20190388561A1 (en) 2019-12-26
JP2023040219A (ja) 2023-03-22
EP3589738A1 (fr) 2020-01-08
IL268946A (en) 2019-10-31
JP2020510433A (ja) 2020-04-09
US11564996B2 (en) 2023-01-31
WO2018160849A8 (fr) 2019-10-03
KR20190125357A (ko) 2019-11-06
RU2019130004A (ru) 2021-04-01
JP7211960B2 (ja) 2023-01-24
KR102545070B1 (ko) 2023-06-19
BR112019017327A2 (pt) 2020-04-14
AU2018228881B2 (en) 2024-01-25
CA3054136A1 (fr) 2018-09-07
AU2018228881A1 (en) 2019-09-12
CN110582572A (zh) 2019-12-17
RU2019130004A3 (fr) 2021-07-05
KR20230093072A (ko) 2023-06-26
US20230233709A1 (en) 2023-07-27
EP3589738A4 (fr) 2021-01-06

Similar Documents

Publication Publication Date Title
US20230323398A1 (en) Viral vectors for the treatment of retinal dystrophy
US20230233709A1 (en) Gene therapy for ocular disorders
US20190343920A1 (en) Aav-mediated gene therapy for nphp5 lca-ciliopathy
JP2023116709A (ja) 眼疾患のための遺伝子療法
CA3029646A1 (fr) Procedes et compositions de traitement de troubles et de maladies impliquant rdh12
WO2018200542A1 (fr) Thérapie génique pour troubles oculaires
US11793887B2 (en) Gene therapy for treating peroxisomal disorders
CA3131923A1 (fr) Therapie genique neuroprotectrice ciblant la voie akt

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18760861

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3054136

Country of ref document: CA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112019017327

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2019547457

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018228881

Country of ref document: AU

Date of ref document: 20180301

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20197027823

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018760861

Country of ref document: EP

Effective date: 20191001

ENP Entry into the national phase

Ref document number: 112019017327

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20190820